WO2020236351A1 - Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme - Google Patents

Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme Download PDF

Info

Publication number
WO2020236351A1
WO2020236351A1 PCT/US2020/028207 US2020028207W WO2020236351A1 WO 2020236351 A1 WO2020236351 A1 WO 2020236351A1 US 2020028207 W US2020028207 W US 2020028207W WO 2020236351 A1 WO2020236351 A1 WO 2020236351A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
cell
gene therapy
transgene
glycoside hydrolase
Prior art date
Application number
PCT/US2020/028207
Other languages
French (fr)
Inventor
Kathrin Christine MEYER
Shibi LIKHITE
Original Assignee
Research Institute At Nationwide Children's Hospital
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Research Institute At Nationwide Children's Hospital filed Critical Research Institute At Nationwide Children's Hospital
Priority to JP2021568543A priority Critical patent/JP2022533645A/en
Priority to EP20723738.9A priority patent/EP3969059A1/en
Priority to CN202080051735.0A priority patent/CN114126667A/en
Priority to CA3141017A priority patent/CA3141017A1/en
Priority to US17/611,972 priority patent/US20220233655A1/en
Priority to KR1020217040926A priority patent/KR20220009427A/en
Priority to AU2020278499A priority patent/AU2020278499A1/en
Publication of WO2020236351A1 publication Critical patent/WO2020236351A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01018Exo-alpha-sialidase (3.2.1.18), i.e. trans-sialidase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01129Endo-alpha-sialidase (3.2.1.129)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14171Demonstrated in vivo effect

Definitions

  • the present disclosure relates to methods of targeting specific cell types within the retina using optimized gene therapy vectors in combination with a glycoside hydrolase enzyme.
  • the disclosure provides gene therapy vectors to specifically target retinal cells and methods of treating visual impairment, retinal degeneration and vision- related disorders.
  • Ocular administration of gene therapy vectors has many advantages due to the well- defined anatomy of the eye.
  • the eye easy accessibility enables rapid and progressive examinations; the relatively enclosed structure and small size of the eye require lower doses of vector for delivery; the blood-retinal barrier prevents the leakage of vectors into systemic circulation, maintaining a relatively immune-privileged environment; and individual or multiple genes primarily or partially involved in particular ocular disorders have been identified.
  • compositions comprising an optimized gene therapy vector that targets specific cell types, such as the specific cells in the retina and a glycoside hydrolase enzyme.
  • optimized gene therapy vectors are useful for delivering a transgene to specific retinal cells using intravitreal delivery in combination with
  • the disclosure provides for methods of treating a vision-related disorder comprising administering the optimized gene therapy vectors using intravitreal delivery in combination with administration of a glycoside hydrolase enzyme.
  • Gene therapy methods that target specific cells types in the retina have advantages for treating vision-loss related diseases.
  • compositions comprising a gene therapy vector and a glycoside hydrolase enzyme.
  • the glycoside hydrolase enzyme is
  • the gene therapy vector is AAV8, AA9 or Anc80.
  • kits for delivering a transgene to a cell of a subject comprising a gene therapy vector and a glycoside hydrolase enzyme.
  • the cell is a retinal cell.
  • the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
  • the gene therapy is AAV8, AA9 or Anc80.
  • the kit comprises instructions for delivering the gene therapy vector and the glycoside hydrolase enzyme to a subject.
  • the disclosure provides for methods of delivering a transgene to a cell in a subject comprising administering to the subject i) a gene therapy vector encoding the transgene and ii) a glycoside hydrolase enzyme.
  • the cell is a retinal cell.
  • the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme, and/or the gene therapy vector is AAV8, AAV9 or Anc80.
  • the disclosure provides for methods wherein the gene therapy vector and/or the glycoside hydrolase enzyme is administered to the subject using local intravenous (IV) delivery, sub-retinal delivery, intravitreal delivery, intracerebroventricular delivery, intraparenchymal delivery or intrathecal (cerebrospinal fluid) delivery.
  • the disclosed methods result in delivering the transgene to all retinal cells including but not limited to bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell and/or amacrine cell. Delivery to retinal cells is exemplified herein, however the disclosed methods, compositions and uses may target any cell type in which glycoside hydrolase enzymes clear the receptors on the cell membrane. Other cell types include muscle cells, nerve cells such as astrocytes, neurons,
  • oligodendrocytes and scwann cells oligodendrocytes and scwann cells.
  • the disclosure also provides for a composition for delivering a transgene to a cell in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene and ii) a glycoside hydrolase enzyme.
  • the cell is a retinal cell.
  • the disclosure provides for a composition for delivering a transgene to a retinal cell in a subject, wherein the composition comprises a gene therapy vector encoding the transgene, wherein the composition is administered with a second composition comprising a glycoside hydrolase enzyme.
  • the composition is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery, intracistermal injection, intracerebroventricular delivery, intramuscular delivery or intrathecal injection.
  • the disclosure also provides for use of a composition for the preparation of a medicament for delivering a transgene to a cell in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene and ii) a glycoside hydrolase enzyme.
  • the cell is a retinal cell.
  • the disclosure provides for use of a gene therapy vector encoding a transgene for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the medicament is administered with a composition comprising a glycoside hydrolase enzyme.
  • the disclosure provides for use of glycoside hydrolase enzyme for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the medicament is administered with a composition comprising a gene therapy vector encoding the transgene.
  • the medicament is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery or intrathecal delivery.
  • the disclosure also provides for methods of treating visual impairment, retinal degeneration or a vision-related disorder in a subject comprising administering to the subject i) a gene therapy vector encoding a transgene and ii) a glycoside hydrolase enzyme.
  • the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme
  • the gene therapy vector is AAV8, AAV9 or Anc80.
  • the disclosure provides for methods wherein the gene therapy vector and/or the glycoside hydrolase enzyme is administered using local intravenous (IV) delivery, sub-retinal delivery, intravitreal delivery, intracerebroventricular delivery, intraparenchymal delivery, intramuscular delivery or intrathecal delivery.
  • IV local intravenous
  • sub-retinal delivery sub-retinal delivery
  • intravitreal delivery intracerebroventricular delivery
  • intraparenchymal delivery intramuscular delivery or intrathecal delivery.
  • the disclosure also provides for compositions for treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises i) a gene therapy vector encoding a transgene and ii) a glycoside hydrolase enzyme.
  • the disclosure provides for a composition for treating a treating visual impairment or a vision- related disorder in a subject, wherein the composition comprises a glycoside hydrolase enzyme, wherein the composition is administered with a second composition comprising a gene therapy vector encoding the transgene.
  • the composition is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery, intracerebroventricular delivery, intramuscular delivery or intrathecal delivery.
  • the disclosure provides for use of a composition for the preparation of a medicament for treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises a gene therapy vector encoding a transgene.
  • the disclosure also provides for use of a gene therapy vector encoding a transgene for the preparation of a medicament for treating visual impairment or a vision-related disorder in a subject, wherein the medicament is administered with a composition comprising a glycoside hydrolase enzyme.
  • the disclosure provides for use of glycoside hydrolase enzyme for the preparation of a medicament for treating a treating visual impairment or a vision-related disorder in a subject, wherein the medicament is administered with a composition comprising a gene therapy vector encoding the transgene.
  • the medicament is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery, intracerebroventricular delivery, intramuscular delivery or intrathecal delivery.
  • the vision-related disorder is Batten disease, congenital cataracts, congenital glaucoma, retinal degeneration, optic atrophy, eye malformations. Strabismus, ocular misalignment, glaucoma, wet age-related macular degeneration, dry age-related macular degeneration, retinitis pigmentosa, choroideremia, Leber congenital amaurosis, Leber’s hereditary optic neuropathy, early onset retinal dystrophy, achromatopsia, x-linked retinoschisis, Usher Syndrome IB, neovascular age-related macular degeneration, Stargardt’s macular degeneration, diabetic macular degeneration, or diabetic macular edema.
  • the vision-related disorder is a CLN Batten disease such as CLN 1 disease, CLN2 disease, CLN3 disease, CLN4 disease, CLN5 disease, CLN6 disease or CLN8 disease.
  • the transgene is a polynucleotide sequence that encodes a polypeptide of interest or is a nucleic acid that inhibits, interferes or silences expression of a gene of interest, such as a siRNA or miRNA.
  • exemplary transgenes are polynucleotides that encode RPE65, RPGR, ORF15, CNGA3, CMH, ND4, PDE6B, ChR2, MERTK, hRS l, hMYOJA, hABCA4, CD59, anti-hVEGF antibody, endostatin-angiostatin, sFLTOl, or sFLT-1.
  • transgenes include siRNA against RTP801, siRNA against VEGFR-1, siRNA against VEGF, or siRNA against ADRB2.
  • the transgene encodes a CLN polypeptide, such as CLN1, CLN2, CLN3, CLN4, CLN5, CLN6 or CLN8.
  • the gene therapy vector and the glycoside hydrolase enzyme are administered simultaneously, or sequentially.
  • the gene therapy vector and the glycoside hydrolase enzyme are administered using the same mode of delivery or administered for each, or using a different mode of delivery for each.
  • the gene therapy vector and the glycoside hydrolase enzyme are administered as a single administration, for example the gene therapy vector and the glycoside hydrolase enzyme are admixed. Alternatively, the gene therapy vector and the glycoside hydrolase enzyme are administered separately. In some
  • the glycoside hydrolase enzyme is administered at least about 30 minutes before administration of the gene therapy vector.
  • the gene therapy vector is AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVRH10,
  • AAVRH74 AAV 11 , AAV 12, AAV13, AAVTT or Anc80, AAV7m8 and their derivatives.
  • the gene therapy vector comprises a CMV promoter, the p546, or the CB promoter.
  • the gene therapy vector and/or glycoside hydrolase enzyme is administered using intrathecal delivery, and the method further comprises placing the subject in the Trendelenburg position after
  • compositions may comprise a non-ionic, low-osmolar contrast agent.
  • the compositions may comprise a non-ionic, low-osmolar contrast agent is selected from the group consisting of iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, ioxilan, and combinations thereof.
  • Figure 1 provides the schematic of the vectors tested in this disclosure.
  • Figure 2 demonstrates retinal delivery of the GFP transgene after intravitreal delivery, and demonstrates that administration in combination with neuraminidase enhanced penetration of the transgene.
  • Figure 3 provides a schematic detailing the timing of intravitreal injection with and without neuraminidase in 8 week wild-type mice.
  • Figures 4A-4C demonstrate that the addition of neuraminidase significantly increases viral transduction of bipolar cells. The tissues were stained for GFP and Otx which is a bipolar cell specific marker.
  • AAV gene therapy for targeting the eye to treat vision-related disordersrequires specific targeting of different cell types.
  • the disclosure provides experimental data comparing different gene therapy vectors, promoters and routes of administration to determine the optimal gene therapy vector for targeting delivery of a transgene to specific cell types in the retina of mice and non-human primates.
  • the data focuses on admini tration of AAV9 and Anc80 vectors, but the disclosure contemplates using any gene therapy vector that comprises a promoter that specifically targets a retinal cell, and these optimized vectors are administered using local intravenous (IV) delivery, sub-retinal delivery, intravitreal delivery, intracerebroventricular delivery, intramuscular delivery, intraparenchymal delivery or intrathecal delivery.
  • IV local intravenous
  • sub-retinal delivery intravitreal delivery
  • intracerebroventricular delivery intramuscular delivery
  • intraparenchymal delivery or intrathecal delivery.
  • intracerebroventricular injection was effective in targeting transgene expression in the bipolar cells of the retina.
  • intrathecal injections can be used to deliver gene therapy vectors to the eye and specifically for delivering gene therapy vectors to bipolar cells.
  • Adeno-associated virus is a replication-deficient parvovirus, the single- stranded DNA genome of which is about 4.7 kb in length including two 145 nucleotide inverted terminal repeats (ITRs) and may be used to refer to the virus itself or derivatives thereof. The term covers all subtypes and both naturally occurring and recombinant forms, except where specified otherwise.
  • ITRs inverted terminal repeats
  • the serotypes of AAV are each associated with a specific clade, the members of which share serologic and functional similarities. Thus, AAVs may also be referred to by the clade.
  • AAV9 sequences are referred to as“clade F” sequences (Gao et ah, J. Virol., 78: 6381-6388 (2004).
  • the present disclosure contemplates the use of any sequence within a specific clade, e.g., clade F.
  • the nucleotide sequences of the genomes of the AAV serotypes are known.
  • the complete genome of AAV-1 is provided in GenBank Accession No.
  • GenBank Accession No. AF085716 the complete genome of AAV-6 is provided in GenBank Accession No. NC_00 1862; at least portions of AAV-7 and AAV-8 genomes are provided in GenBank Accession Nos. AX753246 and AX753249, respectively; the AAV-9 genome is provided in Gao et al., J. Virol., 78: 6381-6388 (2004); the AAV-10 genome is provided in Mol. Ther., 73(1): 67-76 (2006); the AAV-11 genome is provided in Virology, 330(2) ⁇ . 375- 383 (2004); portions of the AAV-12 genome are provided in Genbank Accession No.
  • DQ813647 portions of the AAV-13 genome are provided in Genbank Accession No.
  • Anc80 is an AAV vector that is of AAV1, AAV2, AAV8 and AAV9.
  • the sequence of Anc80 is provided in Zinn et al., Cell Reports 12: 1056-1068, 2015, Vandenberghe et al, PCT/US2014/060163, both of which are incorporated by reference herein, in their entirety and GenBank Accession Nos. KT235804-KT235812.
  • Cis- acting sequences directing viral DNA replication (rep) [0032]
  • encapsidation/packaging and host cell chromosome integration are contained within the ITRs.
  • Three AAV promoters (named p5, pi 9, and p40 for their relative map locations) drive the expression of the two AAV internal open reading frames encoding rep and cap genes.
  • the two rep promoters (p5 and pi 9), coupled with the differential splicing of the single AAV intron (at nucleotides 2107 and 2227), result in the production of four rep proteins (rep 78, rep 68, rep 52, and rep 40) from the rep gene.
  • Rep proteins possess multiple enzymatic properties that are ultimately responsible for replicating the viral genome.
  • the cap gene is expressed from the p40 promoter and it encodes the three capsid proteins VP1, VP2, and VP3. Alternative splicing and non-consensus translational start sites are responsible for the production of the three related capsid proteins.
  • a single consensus polyadenylation site is located at map position 95 of the AAV genome. The life cycle and genetics of AAV are reviewed in Muzyczka, Current Topics in Microbiology and Immunology, 158 97-129 (1992).
  • AAV possesses unique features that make it attractive as a vector for delivering foreign DNA to cells, for example, in gene therapy.
  • AAV infection of cells in culture is noncytopathic, and natural infection of humans and other animals is silent and asymptomatic.
  • AAV infects many am alian cells allowing the possibility of targeting many different tissues in vivo.
  • AAV transduces slowly dividing and non-dividing cells, and can persist essentially for the lifetime of those cells as a transcriptionally active nuclear episome (extrachromosomal element).
  • the native AAV proviral genome is infectious as cloned DNA in plasmids which makes construction of recombinant genomes feasible.
  • the signals directing AAV replication, genome encapsidation and integration are contained within the ITRs of the AAV genome, some or all of the internal approximately 4.3 kb of the genome (encoding replication and structural capsid proteins, rep- cap) may be replaced with foreign DNA such as a gene cassette containing a promoter, a DNA of interest and a polyadenylation signal. In some instances, the rep and cap proteins are provided in trans.
  • AAV is an extremely stable and hearty virus. It easily withstands the conditions used to inactivate adenovirus (56° to 65°C for several hours), making cold preservation of AAV less critical. AAV may even be lyophilized. Finally, AAV-infected cells are not resistant to superinfection.
  • AAV refers to the wild type AAV virus or viral particles.
  • AAV wild type AAV virus or viral particles.
  • AAV a recombinant AAV virus or recombinant infectious
  • rAAV rAAV virus
  • rAAV viral particle rAAV viral particle
  • the term“rAAV genome” refers to a polynucleotide sequence that is derived from a native AAV genome that has been modified. In some embodiments, the rAAV genome has been modified to remove the native cap and rep genes. In some embodiments, the rAAV genome comprises the endogenous 5’ and 3’ inverted terminal repeats (ITRs). In some embodiments, the rAAV genome comprises ITRs from an AAV serotype that is different from the AAV serotype from which the AAV genome was derived. In some embodiments, the rAAV genome comprises a transgene of interest flanked on the 5’ and 3’ ends by inverted terminal repeat (ITR). In some embodiments, the rAAV genome comprises a“gene cassette.”
  • the term“scAAV” refers to a rAAV virus or rAAV viral particle comprising a self complementary genome.
  • the term“ssAAV” refers to a rAAV virus or rAAV viral particle comprising a single- stranded genome.
  • the rAAV genomes provided herein comprise one or more AAV ITRs flanking the transgene polynucleotide sequence.
  • the transgene polynucleotide sequence is operatively linked to transcriptional control elements (including, but not limited to, promoters, enhancers and/or polyadenylation signal sequences) that are functional in target cells to form a gene cassette.
  • promoters are the CMV promoter, chicken b actin promoter (CB), and the P546 promoter. Additional promoters are contemplated herein including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor vims (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia vims promoter, an Epstein-Barr vims immediate early promoter, a Rous sarcoma vims promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the elongation factor- la promoter, the hemoglobin promoter, and the creatine kinase promoter.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor vims
  • HSV human immunodeficiency virus
  • LTR long terminal repeat
  • MoMuLV promoter an
  • CMV promoter sequence a CB promoter sequence, a P546 promoter sequence, and promoter sequences at least: 65%, 70%, 75%,
  • transcription control elements are tissue specific control elements, for example, promoters that allow expression specifically within neurons or specifically within astrocytes. Examples include neuron specific enolase and glial fibrillary acidic protein promoters. Inducible promoters are also contemplated. Non-limiting examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline-regulated promoter.
  • the gene cassette may also include intron sequences to facilitate processing of a transgene RNA transcript when expressed in mammalian cells.
  • intron sequences to facilitate processing of a transgene RNA transcript when expressed in mammalian cells.
  • an intron is the SV40 intron.
  • Packaging refers to a series of intracellular events that result in the assembly and encapsidation of an AAV particle.
  • production refers to the process of producing the rAAV (the infectious, encapsulated rAAV particles) by the packing cells.
  • AAV“rep” and“cap” genes refer to polynucleotide sequences encoding replication and encapsidation proteins, respectively, of adeno-associated virus. AAV rep and cap are referred to herein as AAV“packaging genes.”
  • A“helper virus” for AAV refers to a virus that allows AAV (e.g. wild-type AAV) to be replicated and packaged by a mammalian cell.
  • helper viruses for AAV are known in the art, including adenoviruses, herpesviruses and poxviruses such as vaccinia.
  • the adenovimses may encompass a number of different subgroups, although Adenovirus type 5 of subgroup C is most commonly used. Numerous adenoviruses of human, non-human mammalian and avian origin are known and available from depositories such as the ATCC. Vimses of the herpes family include, for example, herpes simplex viruses (HSV) and Epstein-Barr viruses (EBV), as well as cytomegaloviruses (CMV) and pseudorabies viruses (PRV); which are also available from depositories such as ATCC.
  • HSV herpes simplex viruses
  • EBV Epstein-Barr viruses
  • CMV cytomegaloviruses
  • PRV pseudorabies viruses
  • Helper virus function(s) refers to function(s) encoded in a helper vims genome which allow AAV replication and packaging (in conjunction with other requirements for replication and packaging described herein).
  • helper vims function may be provided in a number of ways, including by providing helper virus or providing, for example, polynucleotide sequences encoding the requisite function(s) to a producer cell in trans.
  • the rAAV genomes provided herein lack AAV rep and cap DNA.
  • AAV DNA in the rAAV genomes (e.g., ITRs) contemplated herein may be from any AAV serotype suitable for deriving a recombinant vims including, but not limited to, AAV serotypes Anc80, AAV- 1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV- 10, AAV-11, AAV-12, AAV-13, AAV rh.74 and AAV-B1.
  • AAV serotypes Anc80 AAV- 1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV- 10, AAV-11, AAV-12, AAV-13, AAV rh.74 and AAV-B1.
  • the nucleotide sequences of the genomes of various AAV serotypes are known in the
  • rAAV with capsid mutations are also contemplated. See, for example, Marsic et al., Molecular Therapy, 22(11): 1900- 1909 (2014).
  • Modified capsids herein are also contemplated and include capsids having various post-translational modifications such as glycosylation and deamidation. Deamidation of asparagine or glutamine side chains resulting in conversion of asparagine residues to aspartic acid or isoaspartic acid residues, and conversion of glutamine to glutamic acid or isoglutamic acid is contemplated in rAAV capsids provided herein. See, for example, Giles et al., Molecular Therapy, 26(12): 2848-2862 (2016). Modified capsids herein are also contemplated to comprise targeting sequences directing the rAAV to the affected tissues and organs requiring treatment.
  • DNA plasmids provided herein comprise rAAV genomes described herein.
  • the DNA plasmids may be transferred to cells permissible for infection with a helper vims of AAV (e.g., adenovirus, El -deleted adenovirus or herpesvirus) for assembly of the rAAV genome into infectious viral particles with AAV9 capsid proteins.
  • AAV e.g., adenovirus, El -deleted adenovirus or herpesvirus
  • rAAV particles require that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from (i.e., not in) the rAAV genome, and helper virus functions.
  • the AAV rep and cap genes may be from any AAV serotype for which recombinant virus can be derived and may be from a different AAV serotype than the rAAV genome ITRs.
  • Production of pseudotyped rAAV is disclosed in, for example, WO 01/83692 which is incorporated by reference herein in its entirety.
  • AAV capsid proteins may be modified to enhance delivery of the recombinant rAAV.
  • capsid proteins are generally known in the art. See, for example, US 2005/0053922 and US 2009/0202490, the disclosures of which are incorporated by reference herein in their entirety.
  • a method of generating a packaging cell is to create a cell line that stably expresses all the necessary components for rAAV production.
  • a plasmid (or multiple plasmids) comprising a rAAV genome lacking AAV rep and cap genes, AAV rep and cap genes separate from the rAAV genome, and a selectable marker, such as a neomycin resistance gene, may be integrated into the genome of a cell.
  • rAAV genomes may be introduced into bacterial plasmids by procedures such as GC tailing (Samulski et ah, 1982, Proc. Natl. Acad. S6.
  • the packaging cell line may then be infected with a helper virus such as adenovirus.
  • a helper virus such as adenovirus.
  • packaging cells that produce infectious rAAV particles.
  • packaging cells may be stably transformed cancer cells such as HeLa cells, 293 cells and PerC.6 cells (a cognate 293 line).
  • packaging cells may be cells that are not transformed cancer cells such as low passage 293 cells (human fetal kidney cells transformed with El of adenovirus), MRC-5 cells (human fetal fibroblasts), WI-38 cells (human fetal fibroblasts), Vero cells (monkey kidney cells) and FRhL-2 cells (rhesus fetal lung cells).
  • rAAV infectious encapsidated rAAV particles
  • the genomes of the rAAV lack AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genomes of the rAAV.
  • the rAAV genome can be a self-complementary (sc) genome.
  • a rAAV with a sc genome is referred to herein as a scAAV.
  • the rAAV genome can be a single-stranded (ss) genome.
  • a rAAV with a single-stranded genome is referred to herein as an ssAAV.
  • the rAAV may be purified by methods standard in the art such as by column chromatography or cesium chloride gradients. Methods for purifying rAAV from helper virus are known in the art and may include methods disclosed in, for example, Clark et al., Hum. Gene Ther., 10(6): 1031-1039 (1999); Schenpp and Clark, Methods Mol. Med., 69: 427-443 (2002); U.S. Patent No. 6,566,118 and WO 98/09657.
  • compositions comprising rAAV are also provided.
  • Compositions comprise a rAAV encoding a CLN6 polypeptide.
  • Compositions may include two or more rAAV encoding different polypeptides of interest.
  • the rAAV is scAAV or ssAAV.
  • Compositions provided herein comprise rAAV and a pharmaceutically acceptable excipient or excipients.
  • Acceptable excipients are nontoxic to recipients and are preferably inert at the dosages and concentrations employed, and include, but are not limited to, buffers such as phosphate [e.g., phosphate-buffered saline (PBS)], citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as
  • polyvinylpyrrolidone amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt forming counterions such as sodium; and/or nonionic surfactants such as Tween, copolymers such as poloxamer 188, pluronics (e.g., Pluronic F68) or polyethylene glycol (PEG).
  • amino acids such as glycine, glutamine, asparagine, arginine or lysine
  • monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins chelating agents such as EDTA
  • sugar alcohols such as mannitol or sorbitol
  • salt forming counterions such as sodium
  • nonionic surfactants such
  • compositions provided herein can comprise a pharmaceutically acceptable aqueous excipient containing a non-ionic, low-osmolar compound such as iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, or ioxilan, where the aqueous excipient containing the non-ionic, low-osmolar compound can have one or more of the following characteristics: about 180 mg I/m L.
  • a non-ionic, low-osmolar compound such as iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, or ioxilan
  • compositions comprise about 20 to 40% non-ionic, low-osmolar compound or about 25% to about 35% non-ionic, low-osmolar compound.
  • An exemplary composition comprises scAAV or rAAV viral particles formulated in 20mM Tris (pH8.0), ImM MgCk, 200mM NaCl, 0.001% poloxamer 188 and about 25% to about 35% non-ionic, low-osmolar compound.
  • Another exemplary composition comprises scAAV formulated in and IX PBS and 0.001% Pluronic F68.
  • Dosages of rAAV to be administered in methods of the disclosure will vary depending, for example, on the particular rAAV, the mode of administration, the time of administration, the treatment goal, the individual, and the cell type(s) being targeted, and may be determined by methods standard in the art. Dosages may be expressed in units of viral genomes (vg).
  • Dosages contemplated herein include about lxlO 7 , lxlO 8 , lxlO 9 ,5xl0 9 , 6 xlO 9 , 7xl0 9 , 8 l0 9 , 9xl0 9 , lxlO 10 , 2xl0 10 , 3xl0 10 , 4xl0 10 , 5xl0 10 , lxlO 11 , about lxlO 12 , about lxlO 13 , about l.lxlO 13 , about 1.2xl0 13 , about 1.3xl0 13 , about 1.5xl0 13 , about 2 xlO 13 , about 2.5 xlO 13 , about 3 x 10 13 , about 3.5 x 10 13 , about 4x 10 13 , about 4.5x 10 13 , about 5 x 10 13 , about 6xl0 13 , about lxlO 14 , about 2 xlO 14 , about
  • Dosages of about lxlO 9 to about 1 xlO 10 , about 5x 10 9 to about 5 xlO 10 , about lxlOio to about lx 10 11 , about lxlO 11 to about lxlO 15 vg, about lxlO 12 to about lxlO 15 vg, about lxlO 12 to about lxlO 14 vg, about lxlO 13 to about 6xl0 14 vg, and about 6xl0 13 to about l.OxlO 14 vg are also contemplated.
  • One dose exemplified herein is 6xl0 13 vg.
  • Another dose exemplified herein is 1.5xl0 13 vg.
  • the retina cells include bipolar cells, rod photoreceptor cells, cone photoreceptor cell, ganglion cell, Mueller glia cells, microglia cells, horizontal cells or amacrine cells.
  • the term“transduction” is used to refer to the administration/delivery of the CLN6 polynucleotide to a target cell either in vivo or in vitro, via a replication-deficient rAAV of the disclosure resulting in expression of a functional polypeptide by the recipient cell.
  • Transduction of cells with rAAV of the disclosure results in sustained expression of polypeptide or RNA encoded by the rAAV.
  • the present disclosure thus provides methods of administering/delivering to a subject rAAV encoding a transgene encoded polypeptide by an intrathecal, local IV delivery, intracerebroventricular, intramuscular, sub-retinal injection, intravitreous delivery or intraparenchymal delivery, or any combination thereof.
  • Intrathecal delivery refers to delivery into the space under the arachnoid membrane of the brain or spinal cord.
  • intrathecal administration is via intracistemal administration.
  • the disclosed methods of delivery any transgene of interest to a retinal cell is a polynucleotide sequence that encodes a polypeptide of interest or is a nucleic acid that inhibits, interferes or silences expression of a gene of interest, such as a siRNA or miRNA.
  • transgenes are polynucleotides that encode RPE65, RPGR, ORF15, CNGA3, CMH, ND4, PDE6B, ChR2, MERTK, hRSl, hMYOJA, hABCA4, CD59, anti- hVEGF antibody, endostatin-angiostatin, sFLTOl, or sFLT-1.
  • the transgene encodes a CLN polypeptide, such as CLN1, CLN2, CLN3, CLN4, CLN5, CLN6 or CLN8.
  • Additional exemplary transgenes include siRNA against RTP801, siRNA against VEGFR-1, siRNA against VEGF, or siRNA against ADRB2.
  • miRNA that are expressed in the retina are contemplated as transgenes to include in the disclosed optimized gene therapy vectors.
  • Examples of miRNA are provided in Karali et al., Nucleic Acids Res. 2016 Feb 29; 44(4): 1525-1540, which is incorporated by reference herein.
  • rAAV genomes provided herein may comprise a polynucleotide encoding a transgene comprising a polynucleotide sequence encoding any one of RPE65, RPGR,
  • polypeptide encoded by the transgene include polypeptides comprising an amino acid sequence that is at least: 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence encoded by the transgene sequence.
  • rAAV genomes provided herein comprise a polynucleotide encoding a CLN polypeptide, such as CLN1, CLN2, CLN3, CLN4, CLN5, CLN6 and CLN8.
  • the polypeptide include polypeptides comprising an amino acid sequence that is at least: 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a CLN polypeptide amino acid sequence, and which encodes a polypeptide with CLN activity (e.g., at least one of increasing clearance of lysosomal auto fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in a patient when treated as compared to, e.g. the patient prior to treatment).
  • CLN activity e.g., at least one of increasing clearance of lysosomal auto fluorescent storage material, reducing lysosomal accumulation of ATP synth
  • rAAV genomes comprise a polynucleotide encoding a CLN polypeptide or a polynucleotide at least: 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence that encodes a polypeptide with CLN activity (e.g., at least one of increasing clearance of lysosomal auto fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in a patient when treated as compared to, e.g. the patient prior to treatment).
  • CLN activity e.g., at least one of increasing clearance of lysosomal auto fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit
  • rAAV genomes provided herein comprise a transgene comprising a polynucleotide sequence that encodes a polypeptide with a desired activity and that hybridizes under stringent conditions to any one of nucleic acid sequence of a known transgene of interest, or the complement thereof.
  • rAAV genomes provided herein comprise a polynucleotide sequence that encodes a polypeptide with CLN activity and that hybridizes under stringent conditions to any one of nucleic acid sequences encoding a CLN polypeptide, or the complement thereof.
  • stringent is used to refer to conditions that are commonly understood in the art as stringent.
  • Hybridization stringency is principally determined by temperature, ionic strength, and the concentration of denaturing agents such as formamide.
  • Examples of stringent conditions for hybridization and washing include but are not limited to 0.015 M sodium chloride, 0.0015 M sodium citrate at 65-68°C or 0.015 M sodium chloride, 0.0015M sodium citrate, and 50% formamide at 42°C. See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, (Cold Spring Harbor, N.Y. 1989).
  • Intrathecal administration is exemplified herein.
  • Intrathecal delivery refers to delivery into the space under the arachnoid membrane of the brain or spinal cord.
  • intrathecal administration is via intracisternal injection or intralumbar injection.
  • These methods include transducing target cells with one or more rAAV described herein.
  • the rAAV viral particle comprising a transgene is administered or delivered the eye, brain and/or spinal cord of a patient.
  • the polynucleotide is delivered to brain. Areas of the brain contemplated for delivery include, but are not limited to, the motor cortex, visual cortex, cerebellum and the brain stem.
  • the polynucleotide is delivered to the spinal cord. In some embodiments, the polynucleotide is delivered to a lower motor neuron.
  • the polynucleotide may be delivered a retinal cell such as a bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell or amacrine cell.
  • the patient is held in the Trendelenburg position (head down position) after administration of the rAAV (e.g., for about 5, about 10, about 15 or about 20 minutes).
  • the patient may be tilted in the head down position at about 1 degree to about 30 degrees, about 15 to about 30 degrees, about 30 to about 60 degrees, about 60 to about 90 degrees, or about 90 to about 180 degrees).
  • a small scleral incision is made, at or posterior to the equator of the eye with a needle, e.g. 30G needle.
  • Virus or vehicle is delivered sub-retinally via the incision, for example, a fine glass pipette attached by tubing to a Hamilton syringe or via 30G needle and Hamilton syringe.
  • sub-retinal administration is carried out by a clinically trained surgeon using methods known in the art.
  • a needle is inserted into the skull and the liquid is injected into the ventricles containing cerebrospinal fluid.
  • the liquid is injected into the ventricles containing cerebrospinal fluid.
  • intracerebroventicular injections are carried out by a clinically trained surgeon using methods known in the art.
  • compositions may comprise a non-ionic, low- osmolar contrast agent.
  • the compositions may comprise a non-ionic, low-osmolar contrast agent is selected from the group consisting of iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, ioxilan, and combinations thereof.
  • the methods provided herein comprise the step of administering an effective dose, or effective multiple doses, of a composition comprising a rAAV provided herein to a subject (e.g., an animal including, but not limited to, a human patient) in need thereof. If the dose is administered prior to development of the symptoms of the vision-related disorder, the administration is prophylactic. If the dose is administered after the development of symptoms of the vision-related disorder, the administration is therapeutic.
  • An effective dose is a dose that alleviates (eliminates or reduces) at least one symptom associated with the vision-related disorder, that slows or prevents progression of the disorder, that diminishes the extent of disorder, that results in remission (partial or total) of disorder, and/or that prolongs survival and/or vision.
  • methods provided herein result in stabilization, reduced progression of vision loss or retinal degeneration, or improvement in vision or macular degeneration.
  • the vision-related disorder is CLN Batten disease
  • methods provided herein result in stabilization, reduced progression, or improvement in one or more of the scales that are used to evaluate progression and/or improvement in CLN Batten-disease, e.g. the Unified Batten Disease Rating System (UBDRS) or the Hamburg Motor and Language Scale.
  • UBDRS assessment scales as described in Marshall et al., Neurology. 2005 65(2):275-279) [including the UBDRS physical one or more of the scales that are used to evaluate progression and/or improvement in CLN Batten-disease, e.g. the Unified Batten Disease Rating System
  • UBDRS Hamburg Motor and Language Scale.
  • the UBDRS assessment scales (as described in Marshall et al., Neurology. 2005 65(2):275-279) [including the UBDRS physical assessment scale, the UBDRS seizure assessment scale, the UBDRS behavioral assessment scale, the UBDRS capability assessment scale, the UBDRS sequence of symptom onset, and the UBDRS Clinical Global Impressions (CGI)]; the Pediatric Quality of Life Scale
  • PEDSQOL scale, motor function, language function, cognitive function, and survival.
  • methods provided herein may result in one or more of the following: reduced or slowed lysosomal accumulation of autofluorescent storage material, reduced or slowed lysosomal accumulation of ATP Synthase Subunit C, reduced or slowed glial activation (astrocytes and/or microglia) activation; reduced or slowed astrocytosis, and showed a reduction or delay in brain volume loss measured by MRI.
  • the disclosure provides for administering an optimized gene therapy in vector in combination with a glycoside hydrolase enzyme, such as neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
  • a glycoside hydrolase enzyme such as neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
  • Exo-glucosidases remove terminal sialic acids from glycan chains. These sialic acids are found on the outermost end of the glycan chains of all cell types and on most secreted proteins.
  • Glycoside hydrolase enzymes are known to clear cell membrane residues, it is thought that this clearance allows for greater viral receptor targeting and cell entry.
  • neuramindase treatment removes N-linked galactosyl residues, thereby enhancing penetration of AAV that use N- 1 inked galactose residues as receptors for entry into the cell, e.g. AAV9 (see Shen et al. J. Biol. Chem. 286: 13532- 13540, 2011). Delivery to retinal cells is exemplified herein, however the disclosed methods, compositions and uses may target any cell type in which glycoside hydrolase enzymes clear the receptors on the cell membrane.
  • Combination therapies comprising the optimized gene therapy vectors are also provided.
  • the terms“combination therapy” and“combination treatment” refer to administration of a disclosed optimized gene therapy vector with an agent that improves targeting to a retina cell such as an enzyme.
  • the methods comprise administering a glycoside hydrolase enzyme to the subject in combination with
  • the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
  • the agents can be administered simultaneously or sequentially with the vector by the any of the routes of administration described herein.
  • Combination as used herein includes either simultaneous treatment or sequential treatment.
  • the gene therapy vector and the glycoside hydrolase enzyme are administered simultaneously using the same mode of administration, or the gene therapy vector and the glycoside hydrolase enzyme are administered simultaneously each using a different mode of administration.
  • the gene therapy vector and the glycoside hydrolase enzyme are administered sequentially using the same mode of administration, or the gene therapy vector and the glycoside hydrolase enzyme are administered sequentially each using a different mode of administration. Combinations of methods described herein with standard medical treatments are specifically contemplated.
  • the optimized gene therapy vector is administered simultaneously with a glycoside hydrolase enzyme, such as neuraminidase.
  • the optimized gene therapy vector is administered immediately before or immediately after a glycoside hydrolase enzyme, such as neuraminidase.
  • the optimized gene therapy vector is administered within about 15 minutes, within about 20 minutes, within about 25 minutes, within about 30 minutes, within about 45 minutes, within 1 hour, within 2 hours, within 3 hours, within 4 hours, within 5 hours, within 6 hours, within 7 hours, within 8 hours, within 9 hours, within 12 hours, within 24 hours, within 36 hours, or within 48 hours of administration of a glycoside hydrolase enzyme, such as neuraminidase.
  • the glycoside hydrolase enzyme is administered before administration of the optimized gene therapy vector or after administration of the optimized gene therapy vector.
  • the optimized gene therapy vector and the glycoside hydrolase enzyme such as neuraminidase
  • the disclosure also provides for administering the combination of the optimized gene therapy vector an the glycoside hydrolase enzyme, such as neuraminidase, in separate intravitreal injections.
  • the glycoside hydrolase enzyme e.g. neuramidase
  • the optimized gene therapy vector an the glycoside hydrolase enzyme may be administered using different modes of administration. Examples
  • a human GFP cDNA clone was obtained from Origene, Rockville, MD. GFP cDNA was further subcloned into a self complementary AAV9 genome or an Anc80 genome under the hybrid chicken b-Actin promoter (CB), the CMV enhancer-promoter, or the P546 promoter and tested in vitro and in vivo.
  • CB hybrid chicken b-Actin promoter
  • P546 promoter the P546 promoter
  • the plasmid construct also included one or more of the CB promoter, an intron such as the simian virus 40 (SV40) chimeric intron and a Bovine Growth Hormone (BGH) polyadenylation signal (BGH PolyA).
  • SV40 simian virus 40
  • BGH Bovine Growth Hormone polyadenylation signal
  • the constructs in Fig. 1 were packaged into either AAV9 genome or the Anc80 genome (referred to collectively as“AAV”).
  • mice were is anesthetized for the intravitreal injection as described above. A small incision was made between the lumbus and sclera with 30G needle. Virus or vehicle is delivered into vitreous space via the incision using a fine glass pipette attached by tubing to a Hamilton syringe or via 30G needle and Hamilton syringe. Before and after the injection, ophthaine and vetropolycin are applied topically, mice are allowed to recover via standard of care (heated cage for recovery, food on the bottom of cage, long sipper tube) and monitored until stable
  • scAAV9.CB.GFP was administered to mice (1 - 5 months old) via one intravitreal injection and expression was monitored at various time points over a course of two months.
  • the AAV and Anc80 were administered at a dose ranging from 9xl0 9 and 3.2xl0 10 vg which was diluted in PBS or straight rAAVinjected.
  • the rAAV was administered simultaneously with neuraminidase or without neuraminidase within a single intravitreal injection.
  • the rAAV was admixed with neuraminidase immediately prior to injection and applied as a single solution.
  • intravitreal injections results in expression of the GFP in the retina.
  • administration of the scAAV9.CB.GFP in combination with neuraminidase enhanced penetration of the transgene into the outer layers of the retina.
  • a key of the retinal staining markers is set out below:
  • Calretinin is a makers for horizontal cells (red stain) and stains the inner nuclear layer of the retina. As shown in Fig. 2B, intrvitreal injection of and AAV9.CB.GFP, delivered the transgene to the horizontal cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
  • Otx2 is a nuclear marker for all bipolar cells (red stain) and stains the inner nuclear layer of the retina.
  • AAV9.CB.GFP delivered the transgene to bipolar cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
  • Pax6 is a is a maker for amacrine cells (red stain) and stains the inner nuclear layer of the retina. As shown in Fig. 2D, intravitreal injection of AAV9.CB.GFP delivered the transgene to the amacrine cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
  • Sox2 is a is a maker for Mueller glia cells (red stain) and stains all the nuclei in the inner layer of the retina.
  • AAV9.CB.GFP delivered the transgene to the Mueller glia cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present disclosure relates to methods of targeting specific cell types within the retina using optimized gene therapy vectors in combination with a glycoside hydrolase enzyme, such as neuraminidase. In particular, the disclosure provides gene therapy vectors administered with a glycoside hydrolase enzyme to specifically target retinal cells and methods of treating visual impairment, retinal degeneration and vision-related disorders.

Description

IMPROVED DELIVERY OF GENE THERAPY VECTORS TO RETINAL CELLS
USING A GLYCOSIDE HYDROLASE ENZYME
[0001] This application claims priority to U.S. Provisional Application No. 62/849,794 filed on May 17, 2019 which is incorporated by reference herein in its entirety.
Incorporation By Reference Of Material Submitted Electronically
[0002] The Sequence Listing, which is a part of the present disclosure, is submitted concurrently with the specification as a text file. The name of the text file containing the Sequence Listing is“ 55603_Seqlisting.txt", which was created on April 13, 2020 and is 15,459 bytes in size. The subject matter of the Sequence Listing is incorporated herein in its entirety by reference.
Field
[0003] The present disclosure relates to methods of targeting specific cell types within the retina using optimized gene therapy vectors in combination with a glycoside hydrolase enzyme. In particular, the disclosure provides gene therapy vectors to specifically target retinal cells and methods of treating visual impairment, retinal degeneration and vision- related disorders.
Background
[0004] Ocular administration of gene therapy vectors has many advantages due to the well- defined anatomy of the eye. In particular, the eye’s easy accessibility enables rapid and progressive examinations; the relatively enclosed structure and small size of the eye require lower doses of vector for delivery; the blood-retinal barrier prevents the leakage of vectors into systemic circulation, maintaining a relatively immune-privileged environment; and individual or multiple genes primarily or partially involved in particular ocular disorders have been identified.
[0005] Ocular administration of gene therapy vectors has shown some promising results. Currently, there are a number of clinical gene therapy trials targeting vision-loss related diseases, and these trials mainly target hereditary retinal disease. For example, clinical trials have investigated Leber congenital amaurosis (LCA), leber's hereditary optic neuropathy and retinitis pigmentosa. To date, AAV vectors, particular AAV2 serotype, have been the most commonly used in ocular gene therapy. See Lee et ah, Progress in retinal and eye research 68: 31-53, 2019 [0006] Neuronal ceroid lipofuscinoses (NCLs) are a group of severe neurodegenerative disorders, which are collectively referred to as Batten disease. These disorders affect the nervous system and typically cause worsening problems with e.g. movement, vision and thinking ability. The different NCLs are distinguished by their genetic cause. Partial or complete loss of vision often develops in patients who have childhood forms of Batten disease. In particular, in people suffering from Batten disease, lipofuscin accumulates inside cells, including those of the brain and retina. The buildup of lipofuscin damages the photoreceptors in the retina, optic nerve, and area of the brain that processes vision.
[0007] Currently, there is a need for improved gene therapy methods that target specific cell types in the retina. Furthermore, there are no therapies that can reverse the symptoms of Batten Disease. Thus, there is a need in the art for treatments for Batten Disease.
Summary
[0008] The disclosure provides compositions comprising an optimized gene therapy vector that targets specific cell types, such as the specific cells in the retina and a glycoside hydrolase enzyme. These optimized gene therapy vectors are useful for delivering a transgene to specific retinal cells using intravitreal delivery in combination with
administration of a glycoside hydrolase enzyme. The administration of the glycoside hydrolase enzyme enhanced gene therapy penetration within the retina. The disclosure provides for methods of treating a vision-related disorder comprising administering the optimized gene therapy vectors using intravitreal delivery in combination with administration of a glycoside hydrolase enzyme. Gene therapy methods that target specific cells types in the retina have advantages for treating vision-loss related diseases.
[0009] The disclosure provides for compositions comprising a gene therapy vector and a glycoside hydrolase enzyme. For example, the glycoside hydrolase enzyme is
neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme. In some embodiments, the gene therapy vector is AAV8, AA9 or Anc80.
[0010] In exemplary embodiments, the disclosure provides for compositions formulated for local intravenous delivery, sub-retinal delivery, intravitreal delivery,
intracerebroventricular or intrathecal delivery. For example, the disclosure provides for compositions in which the gene therapy vector and the glycoside hydrolase enzyme are admixed for administration simultaneously. [0011] In addition, the disclosure provides for kits for delivering a transgene to a cell of a subject comprising a gene therapy vector and a glycoside hydrolase enzyme. For example, the cell is a retinal cell. In exemplary embodiments, the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme. In addition, the gene therapy is AAV8, AA9 or Anc80. Optionally, the kit comprises instructions for delivering the gene therapy vector and the glycoside hydrolase enzyme to a subject.
[0012] The disclosure provides for methods of delivering a transgene to a cell in a subject comprising administering to the subject i) a gene therapy vector encoding the transgene and ii) a glycoside hydrolase enzyme. For example, the cell is a retinal cell. In some
embodiments, the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme, and/or the gene therapy vector is AAV8, AAV9 or Anc80. The disclosure provides for methods wherein the gene therapy vector and/or the glycoside hydrolase enzyme is administered to the subject using local intravenous (IV) delivery, sub-retinal delivery, intravitreal delivery, intracerebroventricular delivery, intraparenchymal delivery or intrathecal (cerebrospinal fluid) delivery. For example, the disclosed methods result in delivering the transgene to all retinal cells including but not limited to bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell and/or amacrine cell. Delivery to retinal cells is exemplified herein, however the disclosed methods, compositions and uses may target any cell type in which glycoside hydrolase enzymes clear the receptors on the cell membrane. Other cell types include muscle cells, nerve cells such as astrocytes, neurons,
oligodendrocytes and scwann cells.
[0013] The disclosure also provides for a composition for delivering a transgene to a cell in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene and ii) a glycoside hydrolase enzyme. For example, the cell is a retinal cell. In additional embodiments, the disclosure provides for a composition for delivering a transgene to a retinal cell in a subject, wherein the composition comprises a gene therapy vector encoding the transgene, wherein the composition is administered with a second composition comprising a glycoside hydrolase enzyme. For example, the composition is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery, intracistermal injection, intracerebroventricular delivery, intramuscular delivery or intrathecal injection. [0014] The disclosure also provides for use of a composition for the preparation of a medicament for delivering a transgene to a cell in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene and ii) a glycoside hydrolase enzyme. For example, the cell is a retinal cell. In some embodiments, the disclosure provides for use of a gene therapy vector encoding a transgene for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the medicament is administered with a composition comprising a glycoside hydrolase enzyme. In other embodiments, the disclosure provides for use of glycoside hydrolase enzyme for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the medicament is administered with a composition comprising a gene therapy vector encoding the transgene. For example, the medicament is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery or intrathecal delivery.
[0015] The disclosure also provides for methods of treating visual impairment, retinal degeneration or a vision-related disorder in a subject comprising administering to the subject i) a gene therapy vector encoding a transgene and ii) a glycoside hydrolase enzyme. In some embodiments, the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme, and/or the gene therapy vector is AAV8, AAV9 or Anc80. The disclosure provides for methods wherein the gene therapy vector and/or the glycoside hydrolase enzyme is administered using local intravenous (IV) delivery, sub-retinal delivery, intravitreal delivery, intracerebroventricular delivery, intraparenchymal delivery, intramuscular delivery or intrathecal delivery.
[0016] The disclosure also provides for compositions for treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises i) a gene therapy vector encoding a transgene and ii) a glycoside hydrolase enzyme. In some embodiments, the disclosure provides for a composition for treating a treating visual impairment or a vision- related disorder in a subject, wherein the composition comprises a glycoside hydrolase enzyme, wherein the composition is administered with a second composition comprising a gene therapy vector encoding the transgene. For example, the composition is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery, intracerebroventricular delivery, intramuscular delivery or intrathecal delivery. [0017] In additional embodiments, the disclosure provides for use of a composition for the preparation of a medicament for treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises a gene therapy vector encoding a transgene. The disclosure also provides for use of a gene therapy vector encoding a transgene for the preparation of a medicament for treating visual impairment or a vision-related disorder in a subject, wherein the medicament is administered with a composition comprising a glycoside hydrolase enzyme. In addition, the disclosure provides for use of glycoside hydrolase enzyme for the preparation of a medicament for treating a treating visual impairment or a vision-related disorder in a subject, wherein the medicament is administered with a composition comprising a gene therapy vector encoding the transgene. For example, the medicament is formulated for administering the gene therapy vector using local intravenous delivery, sub-retinal delivery, intravitreous delivery, intracerebroventricular delivery, intramuscular delivery or intrathecal delivery.
[0018] For example, the vision-related disorder is Batten disease, congenital cataracts, congenital glaucoma, retinal degeneration, optic atrophy, eye malformations. Strabismus, ocular misalignment, glaucoma, wet age-related macular degeneration, dry age-related macular degeneration, retinitis pigmentosa, choroideremia, Leber congenital amaurosis, Leber’s hereditary optic neuropathy, early onset retinal dystrophy, achromatopsia, x-linked retinoschisis, Usher Syndrome IB, neovascular age-related macular degeneration, Stargardt’s macular degeneration, diabetic macular degeneration, or diabetic macular edema. In a particular embodiment, the vision-related disorder is a CLN Batten disease such as CLN 1 disease, CLN2 disease, CLN3 disease, CLN4 disease, CLN5 disease, CLN6 disease or CLN8 disease.
[0019] In any of the disclosed methods, uses or compositions, the transgene is a polynucleotide sequence that encodes a polypeptide of interest or is a nucleic acid that inhibits, interferes or silences expression of a gene of interest, such as a siRNA or miRNA. Exemplary transgenes are polynucleotides that encode RPE65, RPGR, ORF15, CNGA3, CMH, ND4, PDE6B, ChR2, MERTK, hRS l, hMYOJA, hABCA4, CD59, anti-hVEGF antibody, endostatin-angiostatin, sFLTOl, or sFLT-1. Additional exemplary transgenes include siRNA against RTP801, siRNA against VEGFR-1, siRNA against VEGF, or siRNA against ADRB2. In one embodiment, the transgene encodes a CLN polypeptide, such as CLN1, CLN2, CLN3, CLN4, CLN5, CLN6 or CLN8. [0020] In any of the disclosed methods, compositions or uses disclosed herein, the gene therapy vector and the glycoside hydrolase enzyme are administered simultaneously, or sequentially. In addition in any of the disclosed methods, compositions or uses, the gene therapy vector and the glycoside hydrolase enzyme are administered using the same mode of delivery or administered for each, or using a different mode of delivery for each. In any of the disclosed methods, uses or compositions, the gene therapy vector and the glycoside hydrolase enzyme are administered as a single administration, for example the gene therapy vector and the glycoside hydrolase enzyme are admixed. Alternatively, the gene therapy vector and the glycoside hydrolase enzyme are administered separately. In some
embodiments, the glycoside hydrolase enzyme is administered at least about 30 minutes before administration of the gene therapy vector.
[0021] In any of the disclosed methods, uses or compositions, the gene therapy vector is AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVRH10,
AAVRH74, AAV 11 , AAV 12, AAV13, AAVTT or Anc80, AAV7m8 and their derivatives.
[0022] In any of the disclosed methods, uses or compositions, the gene therapy vector comprises a CMV promoter, the p546, or the CB promoter.
[0023] In addition, in any of the disclosed methods, uses or compositions, the gene therapy vector and/or glycoside hydrolase enzyme is administered using intrathecal delivery, and the method further comprises placing the subject in the Trendelenburg position after
administering of the gene therapy vector.
[0024] In any of the methods, uses and compositions provided the compositions may comprise a non-ionic, low-osmolar contrast agent. For example, the compositions may comprise a non-ionic, low-osmolar contrast agent is selected from the group consisting of iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, ioxilan, and combinations thereof.
Brief Description of the Drawings
[0025] Figure 1 provides the schematic of the vectors tested in this disclosure.
[0026] Figure 2 demonstrates retinal delivery of the GFP transgene after intravitreal delivery, and demonstrates that administration in combination with neuraminidase enhanced penetration of the transgene.
[0027] Figure 3 provides a schematic detailing the timing of intravitreal injection with and without neuraminidase in 8 week wild-type mice. [0028] Figures 4A-4C demonstrate that the addition of neuraminidase significantly increases viral transduction of bipolar cells. The tissues were stained for GFP and Otx which is a bipolar cell specific marker.
Detailed Description
[0029] Optimization of AAV gene therapy for targeting the eye to treat vision-related disordersrequires specific targeting of different cell types. The disclosure provides experimental data comparing different gene therapy vectors, promoters and routes of administration to determine the optimal gene therapy vector for targeting delivery of a transgene to specific cell types in the retina of mice and non-human primates.
[0030] The data focuses on admini tration of AAV9 and Anc80 vectors, but the disclosure contemplates using any gene therapy vector that comprises a promoter that specifically targets a retinal cell, and these optimized vectors are administered using local intravenous (IV) delivery, sub-retinal delivery, intravitreal delivery, intracerebroventricular delivery, intramuscular delivery, intraparenchymal delivery or intrathecal delivery. For example, the data demonstrated that AAV9 injected directly into the cerebrospinal fluid via
intracerebroventricular injection was effective in targeting transgene expression in the bipolar cells of the retina. Thus, intrathecal injections can be used to deliver gene therapy vectors to the eye and specifically for delivering gene therapy vectors to bipolar cells.
Gene Therapy Vectors
[0031] Adeno-associated virus (AAV) is a replication-deficient parvovirus, the single- stranded DNA genome of which is about 4.7 kb in length including two 145 nucleotide inverted terminal repeats (ITRs) and may be used to refer to the virus itself or derivatives thereof. The term covers all subtypes and both naturally occurring and recombinant forms, except where specified otherwise. There are multiple serotypes of AAV. The serotypes of AAV are each associated with a specific clade, the members of which share serologic and functional similarities. Thus, AAVs may also be referred to by the clade. For example, AAV9 sequences are referred to as“clade F” sequences (Gao et ah, J. Virol., 78: 6381-6388 (2004). The present disclosure contemplates the use of any sequence within a specific clade, e.g., clade F. The nucleotide sequences of the genomes of the AAV serotypes are known. For example, the complete genome of AAV-1 is provided in GenBank Accession No.
NC_002077; the complete genome of AAV-2 is provided in GenBank Accession No.
NC_001401 and Srivastava et ah, J. Virol., 45: 555-564 (1983); the complete genome of AAV-3 is provided in GenBank Accession No. NC_1829; the complete genome of AAV-4 is provided in GenBank Accession No. NC_001829; the AAV-5 genome is provided in
GenBank Accession No. AF085716; the complete genome of AAV-6 is provided in GenBank Accession No. NC_00 1862; at least portions of AAV-7 and AAV-8 genomes are provided in GenBank Accession Nos. AX753246 and AX753249, respectively; the AAV-9 genome is provided in Gao et al., J. Virol., 78: 6381-6388 (2004); the AAV-10 genome is provided in Mol. Ther., 73(1): 67-76 (2006); the AAV-11 genome is provided in Virology, 330(2)·. 375- 383 (2004); portions of the AAV-12 genome are provided in Genbank Accession No.
DQ813647; portions of the AAV-13 genome are provided in Genbank Accession No.
EU285562. The sequence of the AAV rh.74 genome is provided in see U.S. Patent
9,434,928, incorporated herein by reference. The sequence of the AAV-B 1 genome is provided in Choudhury et al, Mol. Ther., 24(1): 1247-1257 (2016). Anc80 is an AAV vector that is of AAV1, AAV2, AAV8 and AAV9. The sequence of Anc80 is provided in Zinn et al., Cell Reports 12: 1056-1068, 2015, Vandenberghe et al, PCT/US2014/060163, both of which are incorporated by reference herein, in their entirety and GenBank Accession Nos. KT235804-KT235812.
[0032] Cis- acting sequences directing viral DNA replication (rep),
encapsidation/packaging and host cell chromosome integration are contained within the ITRs. Three AAV promoters (named p5, pi 9, and p40 for their relative map locations) drive the expression of the two AAV internal open reading frames encoding rep and cap genes. The two rep promoters (p5 and pi 9), coupled with the differential splicing of the single AAV intron (at nucleotides 2107 and 2227), result in the production of four rep proteins (rep 78, rep 68, rep 52, and rep 40) from the rep gene. Rep proteins possess multiple enzymatic properties that are ultimately responsible for replicating the viral genome. The cap gene is expressed from the p40 promoter and it encodes the three capsid proteins VP1, VP2, and VP3. Alternative splicing and non-consensus translational start sites are responsible for the production of the three related capsid proteins. A single consensus polyadenylation site is located at map position 95 of the AAV genome. The life cycle and genetics of AAV are reviewed in Muzyczka, Current Topics in Microbiology and Immunology, 158 97-129 (1992).
[0033] AAV possesses unique features that make it attractive as a vector for delivering foreign DNA to cells, for example, in gene therapy. AAV infection of cells in culture is noncytopathic, and natural infection of humans and other animals is silent and asymptomatic. Moreover, AAV infects many am alian cells allowing the possibility of targeting many different tissues in vivo. Moreover, AAV transduces slowly dividing and non-dividing cells, and can persist essentially for the lifetime of those cells as a transcriptionally active nuclear episome (extrachromosomal element). The native AAV proviral genome is infectious as cloned DNA in plasmids which makes construction of recombinant genomes feasible.
Furthermore, because the signals directing AAV replication, genome encapsidation and integration are contained within the ITRs of the AAV genome, some or all of the internal approximately 4.3 kb of the genome (encoding replication and structural capsid proteins, rep- cap) may be replaced with foreign DNA such as a gene cassette containing a promoter, a DNA of interest and a polyadenylation signal. In some instances, the rep and cap proteins are provided in trans. Another significant feature of AAV is that it is an extremely stable and hearty virus. It easily withstands the conditions used to inactivate adenovirus (56° to 65°C for several hours), making cold preservation of AAV less critical. AAV may even be lyophilized. Finally, AAV-infected cells are not resistant to superinfection.
[0034] The term“AAV” as used herein refers to the wild type AAV virus or viral particles. The terms“AAV,”“AAV virus,” and“AAV viral particle” are used interchangeably herein. The term“rAAV” refers to a recombinant AAV virus or recombinant infectious,
encapsulated viral particles. The terms“rAAV,”“rAAV virus,” and“rAAV viral particle” are used interchangeably herein.
[0035] The term“rAAV genome” refers to a polynucleotide sequence that is derived from a native AAV genome that has been modified. In some embodiments, the rAAV genome has been modified to remove the native cap and rep genes. In some embodiments, the rAAV genome comprises the endogenous 5’ and 3’ inverted terminal repeats (ITRs). In some embodiments, the rAAV genome comprises ITRs from an AAV serotype that is different from the AAV serotype from which the AAV genome was derived. In some embodiments, the rAAV genome comprises a transgene of interest flanked on the 5’ and 3’ ends by inverted terminal repeat (ITR). In some embodiments, the rAAV genome comprises a“gene cassette.”
[0036] The term“scAAV” refers to a rAAV virus or rAAV viral particle comprising a self complementary genome. The term“ssAAV” refers to a rAAV virus or rAAV viral particle comprising a single- stranded genome. [0037] The rAAV genomes provided herein, in some embodiments, comprise one or more AAV ITRs flanking the transgene polynucleotide sequence. The transgene polynucleotide sequence is operatively linked to transcriptional control elements (including, but not limited to, promoters, enhancers and/or polyadenylation signal sequences) that are functional in target cells to form a gene cassette. Examples of promoters are the CMV promoter, chicken b actin promoter (CB), and the P546 promoter. Additional promoters are contemplated herein including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor vims (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia vims promoter, an Epstein-Barr vims immediate early promoter, a Rous sarcoma vims promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the elongation factor- la promoter, the hemoglobin promoter, and the creatine kinase promoter.
[0038] Additionally provided herein are a CMV promoter sequence, a CB promoter sequence, a P546 promoter sequence, and promoter sequences at least: 65%, 70%, 75%,
80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence of the CMV, CB or P546 sequence which exhibit transcription promoting activity.
[0039] Other examples of transcription control elements are tissue specific control elements, for example, promoters that allow expression specifically within neurons or specifically within astrocytes. Examples include neuron specific enolase and glial fibrillary acidic protein promoters. Inducible promoters are also contemplated. Non-limiting examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline-regulated promoter.
The gene cassette may also include intron sequences to facilitate processing of a transgene RNA transcript when expressed in mammalian cells. One example of such an intron is the SV40 intron.
[0040] “Packaging” refers to a series of intracellular events that result in the assembly and encapsidation of an AAV particle. The term“production” refers to the process of producing the rAAV (the infectious, encapsulated rAAV particles) by the packing cells.
[0041] AAV“rep” and“cap” genes refer to polynucleotide sequences encoding replication and encapsidation proteins, respectively, of adeno-associated virus. AAV rep and cap are referred to herein as AAV“packaging genes.” [0042] A“helper virus” for AAV refers to a virus that allows AAV (e.g. wild-type AAV) to be replicated and packaged by a mammalian cell. A variety of such helper viruses for AAV are known in the art, including adenoviruses, herpesviruses and poxviruses such as vaccinia. The adenovimses may encompass a number of different subgroups, although Adenovirus type 5 of subgroup C is most commonly used. Numerous adenoviruses of human, non-human mammalian and avian origin are known and available from depositories such as the ATCC. Vimses of the herpes family include, for example, herpes simplex viruses (HSV) and Epstein-Barr viruses (EBV), as well as cytomegaloviruses (CMV) and pseudorabies viruses (PRV); which are also available from depositories such as ATCC.
[0043] “Helper virus function(s)” refers to function(s) encoded in a helper vims genome which allow AAV replication and packaging (in conjunction with other requirements for replication and packaging described herein). As described herein,“helper vims function” may be provided in a number of ways, including by providing helper virus or providing, for example, polynucleotide sequences encoding the requisite function(s) to a producer cell in trans.
[0044] The rAAV genomes provided herein lack AAV rep and cap DNA. AAV DNA in the rAAV genomes (e.g., ITRs) contemplated herein may be from any AAV serotype suitable for deriving a recombinant vims including, but not limited to, AAV serotypes Anc80, AAV- 1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV- 10, AAV-11, AAV-12, AAV-13, AAV rh.74 and AAV-B1. As noted above, the nucleotide sequences of the genomes of various AAV serotypes are known in the art. rAAV with capsid mutations, are also contemplated. See, for example, Marsic et al., Molecular Therapy, 22(11): 1900- 1909 (2014). Modified capsids herein are also contemplated and include capsids having various post-translational modifications such as glycosylation and deamidation. Deamidation of asparagine or glutamine side chains resulting in conversion of asparagine residues to aspartic acid or isoaspartic acid residues, and conversion of glutamine to glutamic acid or isoglutamic acid is contemplated in rAAV capsids provided herein. See, for example, Giles et al., Molecular Therapy, 26(12): 2848-2862 (2018). Modified capsids herein are also contemplated to comprise targeting sequences directing the rAAV to the affected tissues and organs requiring treatment.
[0045] DNA plasmids provided herein comprise rAAV genomes described herein. The DNA plasmids may be transferred to cells permissible for infection with a helper vims of AAV (e.g., adenovirus, El -deleted adenovirus or herpesvirus) for assembly of the rAAV genome into infectious viral particles with AAV9 capsid proteins. Techniques to produce rAAV, in which an rAAV genome to be packaged, rep and cap genes, and helper virus functions are provided to a cell are standard in the art. Production of rAAV particles requires that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from (i.e., not in) the rAAV genome, and helper virus functions. The AAV rep and cap genes may be from any AAV serotype for which recombinant virus can be derived and may be from a different AAV serotype than the rAAV genome ITRs. Production of pseudotyped rAAV is disclosed in, for example, WO 01/83692 which is incorporated by reference herein in its entirety. In various embodiments, AAV capsid proteins may be modified to enhance delivery of the recombinant rAAV.
Modifications to capsid proteins are generally known in the art. See, for example, US 2005/0053922 and US 2009/0202490, the disclosures of which are incorporated by reference herein in their entirety.
[0046] A method of generating a packaging cell is to create a cell line that stably expresses all the necessary components for rAAV production. For example, a plasmid (or multiple plasmids) comprising a rAAV genome lacking AAV rep and cap genes, AAV rep and cap genes separate from the rAAV genome, and a selectable marker, such as a neomycin resistance gene, may be integrated into the genome of a cell. rAAV genomes may be introduced into bacterial plasmids by procedures such as GC tailing (Samulski et ah, 1982, Proc. Natl. Acad. S6. USA, 79:2077-2081), addition of synthetic linkers containing restriction endonuclease cleavage sites (Laughlin et ak, 1983, Gene, 23:65-73) or by direct, blunt-end ligation (Senapathy & Carter, 1984, J. Biol. Chem., 259:4661-4666). The packaging cell line may then be infected with a helper virus such as adenovirus. The advantages of this method are that the cells are selectable and are suitable for large-scale production of rAAV. Other non-limiting examples of suitable methods employ adenovirus or baculovirus rather than plasmids to introduce rAAV genomes and/or rep and cap genes into packaging cells.
[0047] General principles of rAAV particle production are reviewed in, for example, Carter, 1992, Current Opinions in Biotechnology, 1533-539; and Muzyczka, 1992, Curr. Topics in Microbial and Immunol., 158:97-129). Various approaches are described in Ratschin et ak, Mol. Cell. Biol. 4:2072 (1984); Hermonat et ak, Proc. Natl. Acad. Sci. USA, 81:6466 (1984); Tratschin et ak, Mol. Cell. Biol. 5:3251 (1985); McLaughlin et ak, J. Virol., 62: 1963 (1988); and Lebkowski et ak, 1988 Mol. Cell. Biol., 7:349 (1988). Samulski et al. (1989, J. Virol., 63:3822-3828); U.S. Patent No. 5,173,414; WO 95/13365 and corresponding U.S. Patent No. 5,658.776 ; WO 95/13392; WO 96/17947; PCT/US98/18600; WO 97/09441 (PCT/US96/14423); WO 97/08298 (PCT/US96/13872); WO 97/21825 (PCT/US96/20777); WO 97/06243 (PCT/FR96/01064); WO 99/11764; Perrin et al. (1995) Vaccine 13:1244- 1250; Paul et al. (1993) Human Gene Therapy 4:609-615; Clark et al. (1996) Gene Therapy 3:1124-1132; U.S. Patent. No. 5,786,211; U.S. Patent No. 5,871,982; and U.S. Patent. No. 6,258,595. The foregoing documents are hereby incorporated by reference in their entirety herein, with particular emphasis on those sections of the documents relating to rAAV particle production.
[0048] Further provided herein are packaging cells that produce infectious rAAV particles. In one embodiment packaging cells may be stably transformed cancer cells such as HeLa cells, 293 cells and PerC.6 cells (a cognate 293 line). In another embodiment, packaging cells may be cells that are not transformed cancer cells such as low passage 293 cells (human fetal kidney cells transformed with El of adenovirus), MRC-5 cells (human fetal fibroblasts), WI-38 cells (human fetal fibroblasts), Vero cells (monkey kidney cells) and FRhL-2 cells (rhesus fetal lung cells).
[0049] Also provided herein are rAAV ( e.g ., infectious encapsidated rAAV particles) comprising a rAAV genome of the disclosure. The genomes of the rAAV lack AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genomes of the rAAV. The rAAV genome can be a self-complementary (sc) genome. A rAAV with a sc genome is referred to herein as a scAAV. The rAAV genome can be a single-stranded (ss) genome. A rAAV with a single-stranded genome is referred to herein as an ssAAV.
[0050] The rAAV may be purified by methods standard in the art such as by column chromatography or cesium chloride gradients. Methods for purifying rAAV from helper virus are known in the art and may include methods disclosed in, for example, Clark et al., Hum. Gene Ther., 10(6): 1031-1039 (1999); Schenpp and Clark, Methods Mol. Med., 69: 427-443 (2002); U.S. Patent No. 6,566,118 and WO 98/09657.
[0051] Compositions comprising rAAV are also provided. Compositions comprise a rAAV encoding a CLN6 polypeptide. Compositions may include two or more rAAV encoding different polypeptides of interest. In some embodiments, the rAAV is scAAV or ssAAV. [0052] Compositions provided herein comprise rAAV and a pharmaceutically acceptable excipient or excipients. Acceptable excipients are nontoxic to recipients and are preferably inert at the dosages and concentrations employed, and include, but are not limited to, buffers such as phosphate [e.g., phosphate-buffered saline (PBS)], citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as
polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt forming counterions such as sodium; and/or nonionic surfactants such as Tween, copolymers such as poloxamer 188, pluronics (e.g., Pluronic F68) or polyethylene glycol (PEG).
Compositions provided herein can comprise a pharmaceutically acceptable aqueous excipient containing a non-ionic, low-osmolar compound such as iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, or ioxilan, where the aqueous excipient containing the non-ionic, low-osmolar compound can have one or more of the following characteristics: about 180 mg I/m L. an osmolality by vapor-pressure osmometry of about 322mOsm/kg water, an osmolarity of about 273mOsm/L, an absolute viscosity of about 2.3cp at 20°C and about 1.5cp at 37°C, and a specific gravity of about 1.164 at 37°C. Exemplary compositions comprise about 20 to 40% non-ionic, low-osmolar compound or about 25% to about 35% non-ionic, low-osmolar compound. An exemplary composition comprises scAAV or rAAV viral particles formulated in 20mM Tris (pH8.0), ImM MgCk, 200mM NaCl, 0.001% poloxamer 188 and about 25% to about 35% non-ionic, low-osmolar compound. Another exemplary composition comprises scAAV formulated in and IX PBS and 0.001% Pluronic F68.
[0053] Dosages of rAAV to be administered in methods of the disclosure will vary depending, for example, on the particular rAAV, the mode of administration, the time of administration, the treatment goal, the individual, and the cell type(s) being targeted, and may be determined by methods standard in the art. Dosages may be expressed in units of viral genomes (vg). Dosages contemplated herein include about lxlO7, lxlO8, lxlO9 ,5xl09, 6 xlO9, 7xl09, 8 l09, 9xl09, lxlO10 , 2xl010, 3xl010, 4xl010, 5xl010, lxlO11, about lxlO12, about lxlO13, about l.lxlO13, about 1.2xl013, about 1.3xl013, about 1.5xl013, about 2 xlO13, about 2.5 xlO13, about 3 x 1013, about 3.5 x 1013, about 4x 1013, about 4.5x 1013, about 5 x 1013, about 6xl013, about lxlO14, about 2 xlO14, about 3 x 1014, about 4x 1014about 5xl014, about lxlO15, to about lxlO16, or more total viral genomes. Dosages of about lxlO9 to about 1 xlO10, about 5x 109 to about 5 xlO10, about lxlOio to about lx 1011, about lxlO11 to about lxlO15 vg, about lxlO12 to about lxlO15 vg, about lxlO12 to about lxlO14 vg, about lxlO13 to about 6xl014 vg, and about 6xl013 to about l.OxlO14 vg are also contemplated. One dose exemplified herein is 6xl013 vg. Another dose exemplified herein is 1.5xl013 vg.
[0054] Methods of transducing target retinal cells with rAAV are provided. The retina cells include bipolar cells, rod photoreceptor cells, cone photoreceptor cell, ganglion cell, Mueller glia cells, microglia cells, horizontal cells or amacrine cells.
[0055] The term“transduction” is used to refer to the administration/delivery of the CLN6 polynucleotide to a target cell either in vivo or in vitro, via a replication-deficient rAAV of the disclosure resulting in expression of a functional polypeptide by the recipient cell.
Transduction of cells with rAAV of the disclosure results in sustained expression of polypeptide or RNA encoded by the rAAV. The present disclosure thus provides methods of administering/delivering to a subject rAAV encoding a transgene encoded polypeptide by an intrathecal, local IV delivery, intracerebroventricular, intramuscular, sub-retinal injection, intravitreous delivery or intraparenchymal delivery, or any combination thereof. Intrathecal delivery refers to delivery into the space under the arachnoid membrane of the brain or spinal cord. In some embodiments, intrathecal administration is via intracistemal administration.
Transgenes
[0056] The disclosed methods of delivery any transgene of interest to a retinal cell. The transgene is a polynucleotide sequence that encodes a polypeptide of interest or is a nucleic acid that inhibits, interferes or silences expression of a gene of interest, such as a siRNA or miRNA.
[0057] Exemplary transgenes are polynucleotides that encode RPE65, RPGR, ORF15, CNGA3, CMH, ND4, PDE6B, ChR2, MERTK, hRSl, hMYOJA, hABCA4, CD59, anti- hVEGF antibody, endostatin-angiostatin, sFLTOl, or sFLT-1. In one embodiment, the transgene encodes a CLN polypeptide, such as CLN1, CLN2, CLN3, CLN4, CLN5, CLN6 or CLN8. Additional exemplary transgenes include siRNA against RTP801, siRNA against VEGFR-1, siRNA against VEGF, or siRNA against ADRB2.
[0058] miRNA that are expressed in the retina are contemplated as transgenes to include in the disclosed optimized gene therapy vectors. Examples of miRNA are provided in Karali et al., Nucleic Acids Res. 2016 Feb 29; 44(4): 1525-1540, which is incorporated by reference herein.
[0059] rAAV genomes provided herein may comprise a polynucleotide encoding a transgene comprising a polynucleotide sequence encoding any one of RPE65, RPGR,
ORF15, CNGA3, CMH, ND4, PDE6B, ChR2, MERTK, hRS l, hMYOJA, hABCA4, CD59, PEDF, endostatin-angiostatin genes, sFLT-1, gene encoding an anti-hVEGF antibody. For example, the polypeptide encoded by the transgene include polypeptides comprising an amino acid sequence that is at least: 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence encoded by the transgene sequence.
[0060] rAAV genomes provided herein comprise a polynucleotide encoding a CLN polypeptide, such as CLN1, CLN2, CLN3, CLN4, CLN5, CLN6 and CLN8. The polypeptide include polypeptides comprising an amino acid sequence that is at least: 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a CLN polypeptide amino acid sequence, and which encodes a polypeptide with CLN activity (e.g., at least one of increasing clearance of lysosomal auto fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in a patient when treated as compared to, e.g. the patient prior to treatment).
[0061] rAAV genomes provided herein, in some cases, comprise a polynucleotide encoding a CLN polypeptide or a polynucleotide at least: 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence that encodes a polypeptide with CLN activity (e.g., at least one of increasing clearance of lysosomal auto fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in a patient when treated as compared to, e.g. the patient prior to treatment).
[0062] rAAV genomes provided herein, in some embodiments, comprise a transgene comprising a polynucleotide sequence that encodes a polypeptide with a desired activity and that hybridizes under stringent conditions to any one of nucleic acid sequence of a known transgene of interest, or the complement thereof. In other embodiments, rAAV genomes provided herein comprise a polynucleotide sequence that encodes a polypeptide with CLN activity and that hybridizes under stringent conditions to any one of nucleic acid sequences encoding a CLN polypeptide, or the complement thereof. [0063] The following outlines the disease characteristics of each Batten Disease subtype with emphasis on visual components. The data included in the“Primary Affected Retinal Cell” column was determined based on single cell RNA data compiled from mouse retina. Investigation of this data is still ongoing.
Figure imgf000019_0001
[0064] The term“stringent” is used to refer to conditions that are commonly understood in the art as stringent. Hybridization stringency is principally determined by temperature, ionic strength, and the concentration of denaturing agents such as formamide. Examples of stringent conditions for hybridization and washing include but are not limited to 0.015 M sodium chloride, 0.0015 M sodium citrate at 65-68°C or 0.015 M sodium chloride, 0.0015M sodium citrate, and 50% formamide at 42°C. See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, (Cold Spring Harbor, N.Y. 1989).
Methods of Administration
[0065] Intrathecal administration is exemplified herein. Intrathecal delivery refers to delivery into the space under the arachnoid membrane of the brain or spinal cord. In some embodiments, intrathecal administration is via intracisternal injection or intralumbar injection. These methods include transducing target cells with one or more rAAV described herein. In some embodiments, the rAAV viral particle comprising a transgene is administered or delivered the eye, brain and/or spinal cord of a patient. In some embodiments, the polynucleotide is delivered to brain. Areas of the brain contemplated for delivery include, but are not limited to, the motor cortex, visual cortex, cerebellum and the brain stem. In some embodiments, the polynucleotide is delivered to the spinal cord. In some embodiments, the polynucleotide is delivered to a lower motor neuron. The polynucleotide may be delivered a retinal cell such as a bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell or amacrine cell.
[0066] In some embodiments of methods provided herein, the patient is held in the Trendelenburg position (head down position) after administration of the rAAV (e.g., for about 5, about 10, about 15 or about 20 minutes). For example, the patient may be tilted in the head down position at about 1 degree to about 30 degrees, about 15 to about 30 degrees, about 30 to about 60 degrees, about 60 to about 90 degrees, or about 90 to about 180 degrees).
[0067] For sub-retinal administration, a small scleral incision is made, at or posterior to the equator of the eye with a needle, e.g. 30G needle. Virus or vehicle is delivered sub-retinally via the incision, for example, a fine glass pipette attached by tubing to a Hamilton syringe or via 30G needle and Hamilton syringe. For example, sub-retinal administration is carried out by a clinically trained surgeon using methods known in the art.
[0068] For intracerebroventicular injections, a needle is inserted into the skull and the liquid is injected into the ventricles containing cerebrospinal fluid. For example,
intracerebroventicular injections are carried out by a clinically trained surgeon using methods known in the art.
[0069] In any of the methods of administration, compositions may comprise a non-ionic, low- osmolar contrast agent. For example, the compositions may comprise a non-ionic, low-osmolar contrast agent is selected from the group consisting of iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, ioxilan, and combinations thereof.
[0070] The methods provided herein comprise the step of administering an effective dose, or effective multiple doses, of a composition comprising a rAAV provided herein to a subject (e.g., an animal including, but not limited to, a human patient) in need thereof. If the dose is administered prior to development of the symptoms of the vision-related disorder, the administration is prophylactic. If the dose is administered after the development of symptoms of the vision-related disorder, the administration is therapeutic. An effective dose is a dose that alleviates (eliminates or reduces) at least one symptom associated with the vision-related disorder, that slows or prevents progression of the disorder, that diminishes the extent of disorder, that results in remission (partial or total) of disorder, and/or that prolongs survival and/or vision. In comparison to the subject before treatment or in comparison to an untreated subject, methods provided herein result in stabilization, reduced progression of vision loss or retinal degeneration, or improvement in vision or macular degeneration.
[0071] When the vision-related disorder is CLN Batten disease, comparison to the subject before treatment or in comparison to an untreated subject, methods provided herein result in stabilization, reduced progression, or improvement in one or more of the scales that are used to evaluate progression and/or improvement in CLN Batten-disease, e.g. the Unified Batten Disease Rating System (UBDRS) or the Hamburg Motor and Language Scale. The UBDRS assessment scales (as described in Marshall et al., Neurology. 2005 65(2):275-279) [including the UBDRS physical one or more of the scales that are used to evaluate progression and/or improvement in CLN Batten-disease, e.g. the Unified Batten Disease Rating System
(UBDRS) or the Hamburg Motor and Language Scale. The UBDRS assessment scales (as described in Marshall et al., Neurology. 2005 65(2):275-279) [including the UBDRS physical assessment scale, the UBDRS seizure assessment scale, the UBDRS behavioral assessment scale, the UBDRS capability assessment scale, the UBDRS sequence of symptom onset, and the UBDRS Clinical Global Impressions (CGI)]; the Pediatric Quality of Life Scale
(PEDSQOL) scale, motor function, language function, cognitive function, and survival. In comparison to the subject before treatment or in comparison to an untreated subject, methods provided herein may result in one or more of the following: reduced or slowed lysosomal accumulation of autofluorescent storage material, reduced or slowed lysosomal accumulation of ATP Synthase Subunit C, reduced or slowed glial activation (astrocytes and/or microglia) activation; reduced or slowed astrocytosis, and showed a reduction or delay in brain volume loss measured by MRI.
Glycoside Hydrolase Enzyme
[0072] The disclosure provides for administering an optimized gene therapy in vector in combination with a glycoside hydrolase enzyme, such as neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme. Exo-glucosidases remove terminal sialic acids from glycan chains. These sialic acids are found on the outermost end of the glycan chains of all cell types and on most secreted proteins. Glycoside hydrolase enzymes are known to clear cell membrane residues, it is thought that this clearance allows for greater viral receptor targeting and cell entry. For example, neuramindase treatment removes N-linked galactosyl residues, thereby enhancing penetration of AAV that use N- 1 inked galactose residues as receptors for entry into the cell, e.g. AAV9 (see Shen et al. J. Biol. Chem. 286: 13532- 13540, 2011). Delivery to retinal cells is exemplified herein, however the disclosed methods, compositions and uses may target any cell type in which glycoside hydrolase enzymes clear the receptors on the cell membrane.
[0073] Combination therapies comprising the optimized gene therapy vectors are also provided. The terms“combination therapy” and“combination treatment” refer to administration of a disclosed optimized gene therapy vector with an agent that improves targeting to a retina cell such as an enzyme. In some embodiments, the methods comprise administering a glycoside hydrolase enzyme to the subject in combination with
administration of the gene therapy vector. For example, the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme. The agents can be administered simultaneously or sequentially with the vector by the any of the routes of administration described herein. [0074] Combination as used herein includes either simultaneous treatment or sequential treatment. For example, the gene therapy vector and the glycoside hydrolase enzyme are administered simultaneously using the same mode of administration, or the gene therapy vector and the glycoside hydrolase enzyme are administered simultaneously each using a different mode of administration. In an additional example, the gene therapy vector and the glycoside hydrolase enzyme are administered sequentially using the same mode of administration, or the gene therapy vector and the glycoside hydrolase enzyme are administered sequentially each using a different mode of administration. Combinations of methods described herein with standard medical treatments are specifically contemplated.
[0075] In some embodiments, the optimized gene therapy vector is administered simultaneously with a glycoside hydrolase enzyme, such as neuraminidase. In other embodiments, the optimized gene therapy vector is administered immediately before or immediately after a glycoside hydrolase enzyme, such as neuraminidase. In other embodiments, the optimized gene therapy vector is administered within about 15 minutes, within about 20 minutes, within about 25 minutes, within about 30 minutes, within about 45 minutes, within 1 hour, within 2 hours, within 3 hours, within 4 hours, within 5 hours, within 6 hours, within 7 hours, within 8 hours, within 9 hours, within 12 hours, within 24 hours, within 36 hours, or within 48 hours of administration of a glycoside hydrolase enzyme, such as neuraminidase. In some embodiments, the glycoside hydrolase enzyme is administered before administration of the optimized gene therapy vector or after administration of the optimized gene therapy vector.
[0076] In some embodiments, the optimized gene therapy vector and the glycoside hydrolase enzyme, such as neuraminidase, is administered in a single intravitreal injection. However, the disclosure also provides for administering the combination of the optimized gene therapy vector an the glycoside hydrolase enzyme, such as neuraminidase, in separate intravitreal injections. For example, the glycoside hydrolase enzyme, e.g. neuramidase, is administered about 30 minutes before the administration of the optimized gene therapy vector. In addition, the optimized gene therapy vector an the glycoside hydrolase enzyme may be administered using different modes of administration. Examples
[0077] While the following examples describe specific embodiments, it is understood that variations and modifications will occur to those skilled in the art. Accordingly, only such limitations as appear in the claims should be placed on the invention.
Example 1
Production of scAAV9.GFP and Anc80.GFP
[0078] A human GFP cDNA clone was obtained from Origene, Rockville, MD. GFP cDNA was further subcloned into a self complementary AAV9 genome or an Anc80 genome under the hybrid chicken b-Actin promoter (CB), the CMV enhancer-promoter, or the P546 promoter and tested in vitro and in vivo. A schematic of the plasmid constructs showing the GFP cDNA inserted between AAV2 ITRs is provided in Fig. 1. The plasmid construct also included one or more of the CB promoter, an intron such as the simian virus 40 (SV40) chimeric intron and a Bovine Growth Hormone (BGH) polyadenylation signal (BGH PolyA). The constructs in Fig. 1 were packaged into either AAV9 genome or the Anc80 genome (referred to collectively as“AAV”).
Example 2
Intavitreal Injection of scAAV9.CB.GFP in Combination with Neuraminidase
[0079] The mouse was is anesthetized for the intravitreal injection as described above. A small incision was made between the lumbus and sclera with 30G needle. Virus or vehicle is delivered into vitreous space via the incision using a fine glass pipette attached by tubing to a Hamilton syringe or via 30G needle and Hamilton syringe. Before and after the injection, ophthaine and vetropolycin are applied topically, mice are allowed to recover via standard of care (heated cage for recovery, food on the bottom of cage, long sipper tube) and monitored until stable
[0080] scAAV9.CB.GFP was administered to mice (1 - 5 months old) via one intravitreal injection and expression was monitored at various time points over a course of two months. The AAV and Anc80 were administered at a dose ranging from 9xl09 and 3.2xl010 vg which was diluted in PBS or straight rAAVinjected. In addition, the rAAV was administered simultaneously with neuraminidase or without neuraminidase within a single intravitreal injection. For the simultaneous administration, the rAAV was admixed with neuraminidase immediately prior to injection and applied as a single solution. As shown in Fig. 2A, intravitreal injections results in expression of the GFP in the retina. Furthermore, administration of the scAAV9.CB.GFP in combination with neuraminidase enhanced penetration of the transgene into the outer layers of the retina. A key of the retinal staining markers is set out below:
Figure imgf000025_0001
[0081] Calretinin is a makers for horizontal cells (red stain) and stains the inner nuclear layer of the retina. As shown in Fig. 2B, intrvitreal injection of and AAV9.CB.GFP, delivered the transgene to the horizontal cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
[0082] Otx2 is a nuclear marker for all bipolar cells (red stain) and stains the inner nuclear layer of the retina. As shown in Fig. 2C, intravitreal injection of AAV9.CB.GFP, delivered the transgene to bipolar cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
[0083] Pax6 is a is a maker for amacrine cells (red stain) and stains the inner nuclear layer of the retina. As shown in Fig. 2D, intravitreal injection of AAV9.CB.GFP delivered the transgene to the amacrine cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
[0084] Sox2 is a is a maker for Mueller glia cells (red stain) and stains all the nuclei in the inner layer of the retina. As shown in Fig. 2E, intravitreal injection of AAV9.CB.GFP delivered the transgene to the Mueller glia cells, and neuraminidase enhanced penetration of the transgene into the inner nuclear layer of the retina.
Example 3
[0085] To further investigate the effect of neuraminidase on transduction of 8 week old wild type mice received intravitreal injections of AAV9.CB.GFP with or without neuraminidase as depicted in Figure 3. The ~2xl010 vg of AAV9.CB.GFP was injected in the mice as described in detail in Example 2. The retinal tissue in Figures 4A and Figure 4B was stained for transgene GFP and the bipolar cell specific marker Otx2. The addition of neuraminidase either before or after intravitreal injection of the AAV vector significantly increased viral transduction of the biopolar cells (See Figure 4C).
[0086] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments described herein may be employed. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
[0087] All documents referred to in this application are hereby incorporated by reference in their entirety.
References
[0088] Arsov, T., Smith, K. R., Damiano, J., Franceschetti, S., Canafoglia, L., Bromhead, C. J., ... Berkovic, S. F. (2011). Kufs disease, the major adult form of neuronal ceroid lipofuscinosis, caused by mutations in CLN6. American Journal of Human Genetics, 88(5), 566-573. https://doi.Org/10.1016/j.ajhg.2011.04.004
[0089] Bras, J., Verloes, A., Schneider, S. A., Mole, S. E., & Guerreiro, R. J. (2012).
Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis. Human Molecular Genetics, 27(12), 2646-2650. https://doi.org/10.1093/hmg/dds089
[0090] Canafoglia, L., Morbin, M., Scaioli, V., Pareyson, D., D’Incerti, L., Fugnanesi, V., ... Franceschetti, S. (2014). Recurrent generalized seizures, visual loss, and palinopsia as phenotypic features of neuronal ceroid lipofuscinosis due to progranulin gene mutation. Epilepsia, 55(6), e56-e59. https://doi.org/10.l l l l/epi.12632
[0091] Cannelli, N., Cassandrini, D., Bertini, E., Striano, P., Fusco, L., Gaggero, R., ... Santorelli, F. M. (2006). Novel mutations in CLN8 in Italian variant late infantile neuronal ceroid lipofuscinosis: another genetic hit in the Mediterranean. Neurogenetics, 7(2), 111-117. https://doi.org/10.1007/sl0048-005-0024-y
[0092] Estrada-Cuzcano, A., Martin, S., Chamova, T., Synofzik, M., Timmann, D., Holemans, T., ... Schiile, R. (2017). Loss-of-function mutations in the ATP13A2/PARK9 gene cause complicated hereditary spastic paraplegia (SPG78). Brain : A Journal of
Neurology, 140(2), 287-305. https://doi.org/10.1093/brain/aww307
[0093] Kniffin, C. (2016). OMIM Clinical Synopsis - %609055 - CEROID
LIPOFUSCINOSIS, NEURONAL, 9; CLN9. Retrieved March 19, 2019, from
http ://omim. org/clinicalS ynop sis/609055
[0094] Moen, M. N., Fjrer, R., Hamdani, E. H., Laerdahl, J. K., Menchini, R. J., Vigeland, M. D., ... Chaudhry, F. A. (2016). Pathogenic variants in KCTD7 perturb neuronal K + fluxes and glutamine transport. Brain, 139(12), 3109-3120. https://doi.org/10.1093/brain/aww244
[0095] Mole, S. E., & Williams, R. E. (1993). Neuronal Ceroid-Lipofuscinoses.
GeneReviews®. University of Washington, Seattle. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/20301601
[0096] Noskova, L., Stranecky, V., Hartmannova, H., Pristoupilova, A., Baresova, V., Ivanek, R., ... Kmoch, S. (2011). Mutations in DNAJC5, encoding cysteine-string protein alpha, cause autosomal-dominant adult-onset neuronal ceroid lipofuscinosis. American Journal of Human Genetics, §9(2), 241-252. https://doi.Org/10.1016/j.ajhg.2011.07.003
[0097] Roosing, S., van den Born, L. I., Sangermano, R., Banfi, S., Koenekoop, R. K., Zonneveld-Vrieling, M. N., ... Hoyng, C. B. (2015). Mutations in MFSD8, Encoding a Lysosomal Membrane Protein, Are Associated with Nonsyndromic Autosomal Recessive Macular Dystrophy. Ophthalmology, 722(1), 170-179.
https://doi.Org/10.1016/j.ophtha.2014.07.040
[0098] Staropoli, J. F., Karaa, A., Lim, E. T., Kirby, A., Elbalalesy, N., Romansky, S. G., ... Cotman, S. L. (2012). A homozygous mutation in KCTD7 links neuronal ceroid lipofuscinosis to the ubiquitin-proteasome system. American Journal of Human Genetics, 97(1), 202-208. https://doi.Org/10.1016/j.ajhg.2012.05.023
[0099] Weizmann Institute of Science. (2019). Ceroid Lipofuscinosis, Neuronal, 10 disease: Malacards - Research Articles, Drugs, Genes, Clinical Trials. Retrieved March 19, 2019, from https://www.malacards.org/card/ceroid_lipofuscinosis_neuronal_10_2
[00100] Xin, W., Mullen, T. E„ Kiely, R., Min, J„ Feng, X., Cao, Y„ ... Sims, K. (2010). CLN5 mutations are frequent in juvenile and late-onset non-Finnish patients with NCL. Neurology, 74(1), 565-571. https://doi.org/10.1212/WNL.0b013e3181cff70d
CLN3 nucleotide sequence (SEQ ID NO: 1)
atgggaggct gtgcaggctc gcggcggcgc ttttcggatt ccgaggggga ggagaccgtc 60 ccggagcccc ggctccctct gttggaccat cagggcgcgc attggaagaa cgcggtgggc 120 ttctggctgc tgggcctttg caacaacttc tcttatgtgg tgatgctgag tgccgcccac 180 gacatcctta gccacaagag gacatcggga aaccagagcc atgtggaccc aggcccaacg 240 ccgatccccc acaacagctc atcacgattt gactgcaact ctgtctctac ggctgctgtg 300 ctcctggcgg acatcctccc cacactcgtc atcaaattgt tggctcctct tggccttcac 360 ctgctgccct acagcccccg ggttctcgtc agtgggattt gtgctgctgg aagcttcgtc 420 ctggttgcct tttctcattc tgtggggacc agcctgtgtg gtgtggtctt cgctagcatc 480 tcatcaggcc ttggggaggt caccttcctc tccctcactg ccttctaccc cagggccgtg 540 atctcctggt ggtcctcagg gactggggga gctgggctgc tgggggccct gtcctacctg 600 ggcctcaccc aggccggcct ctcccctcag cagaccctgc tgtccatgct gggtatccct 660 gccctgctgc tggccagcta tttcttgttg ctcacatctc ctgaggccca ggaccctgga 720 ggggaagaag aagcagagag cgcagcccgg cagcccctca taagaaccga ggccccggag 780 tcgaagccag gctccagctc cagcctctcc cttcgggaaa ggtggacagt gttcaagggt 840 ctgctgtggt acattgttcc cttggtcgta gtttactttg ccgagtattt cattaaccag 900 ggactttttg aactcctctt tttctggaac acttccctga gtcacgctca gcaataccgc 960 tggtaccaga tgctgtacca ggctggcgtc tttgcctccc gctcttctct ccgctgctgt 1020 cgcatccgtt tcacctgggc cctggccctg ctgcagtgcc tcaacctggt gttcctgctg 1080 gcagacgtgt ggttcggctt tctgccaagc atctacctcg tcttcctgat cattctgtat 1140 gaggggctcc tgggaggcgc agcctacgtg aacaccttcc acaacatcgc cctggagacc 1200 agtgatgagc accgggagtt tgcaatggcg gccacctgca tctctgacac actggggatc 1260 tccctgtcgg ggctcctggc tttgcctctg catgacttcc tctgccagct ctcctga 1317
CLN3 amino acid sequence (SEQ ID NO: 21
Met Gly Gly Cys Ala Gly Ser Arg Arg Arg Phe Ser Asp Ser Glu Gly
1 5 10 15
Glu Glu Thr Val Pro Glu Pro Arg Leu Pro Leu Leu Asp His Gin Gly
20 25 30
Ala His Trp Lys Asn Ala Val Gly Phe Trp Leu Leu Gly Leu Cys Asn
35 40 45
Asn Phe Ser Tyr Val Val Met Leu Ser Ala Ala His Asp lie Leu Ser
50 55 60
His Lys Arg Thr Ser Gly Asn Gin Ser His Val Asp Pro Gly Pro Thr
65 70 75 80
Pro lie Pro His Asn Ser Ser Ser Arg Phe Asp Cys Asn Ser Val Ser
85 90 95
Thr Ala Ala Val Leu Leu Ala Asp He Leu Pro Thr Leu Val He Lys
100 105 110
Leu Leu Ala Pro Leu Gly Leu His Leu Leu Pro Tyr Ser Pro Arg Val
115 120 125
Leu Val Ser Gly He Cys Ala Ala Gly Ser Phe Val Leu Val Ala Phe
130 135 140
Ser His Ser Val Gly Thr Ser Leu Cys Gly Val Val Phe Ala Ser He
145 150 155 160
Ser Ser Gly Leu Gly Glu Val Thr Phe Leu Ser Leu Thr Ala Phe Tyr
165 170 175
Pro Arg Ala Val He Ser Trp Trp Ser Ser Gly Thr Gly Gly Ala Gly
180 185 190 Leu Leu Gly Ala Leu Ser Tyr Leu Gly Leu Thr Gin Ala Gly Leu Ser
195 200 205
Pro Gin Gin Thr Leu Leu Ser Met Leu Gly lie Pro Ala Leu Leu Leu
210 215 220
Ala Ser Tyr Phe Leu Leu Leu Thr Ser Pro Glu Ala Gin Asp Pro Gly
225 230 235 240
Gly Glu Glu Glu Ala Glu Ser Ala Ala Arg Gin Pro Leu lie Arg Thr
245 250 255
Glu Ala Pro Glu Ser Lys Pro Gly Ser Ser Ser Ser Leu Ser Leu Arg
260 265 270
Glu Arg Trp Thr Val Phe Lys Gly Leu Leu Trp Tyr He Val Pro Leu
275 280 285
Val Val Val Tyr Phe Ala Glu Tyr Phe He Asn Gin Gly Leu Phe Glu
290 295 300
Leu Leu Phe Phe Trp Asn Thr Ser Leu Ser His Ala Gin Gin Tyr Arg
305 310 315 320
Trp Tyr Gin Met Leu Tyr Gin Ala Gly Val Phe Ala Ser Arg Ser Ser
325 330 335
Leu Arg Cys Cys Arg He Arg Phe Thr Trp Ala Leu Ala Leu Leu Gin
340 345 350
Cys Leu Asn Leu Val Phe Leu Leu Ala Asp Val Trp Phe Gly Phe Leu
355 360 365
Pro Ser He Tyr Leu Val Phe Leu He He Leu Tyr Glu Gly Leu Leu
370 375 380
Gly Gly Ala Ala Tyr Val Asn Thr Phe His Asn He Ala Leu Glu Thr
385 390 395 400
Ser Asp Glu His Arg Glu Phe Ala Met Ala Ala Thr Cys He Ser Asp
405 410 415
Thr Leu Gly He Ser Leu Ser Gly Leu Leu Ala Leu Pro Leu His Asp
420 425 430
Phe Leu Cys Gin Leu Ser
435
CLN6 nucleotide sequence (SEQ ID NO: 3)
atggaggcga cgcggaggcg gcagcacctg ggagcgacgg gcggcccagg cgcgcagctg 60 ggcgcctcct tcctgcaggc caggcatggc tctgtgagcg ctgatgaggc tgcccgcacg 120 gctcccttcc acctcgacct ctggttctac ttcacactgc agaactgggt tctggacttt 180 gggcgtccca ttgccatgct ggtattccct ctcgagtggt ttccactcaa caagcccagt 240 gttggggact acttccacat ggcctacaac gtcatcacgc cctttctctt gctcaagctc 300 atcgagcggt ccccccgcac cctgccacgc tccatcacgt acgtgagcat catcatcttc 360 atcatgggtg ccagcatcca cctggtgggt gactctgtca accaccgcct gctcttcagt 420 ggctaccagc accacctgtc tgtccgtgag aaccccatca tcaagaatct caagccggag 480 acgctgatcg actcctttga gctgctctac tattatgatg agtacctggg tcactgcatg 540 tggtacatcc ccttcttcct catcctcttc atgtacttca gcggctgctt tactgcctct 600 aaagctgaga gcttgattcc agggcctgcc ctgctcctgg tggcacccag tggcctgtac 660 tactggtacc tggtcaccga gggccagatc ttcatcctct tcatcttcac cttcttcgcc 720 atgctggccc tcgtcctgca ccagaagcgc aagcgcctct tcctggacag caacggcctc 780 ttcctcttct cctccttcgc actgaccctc ttgcttgtgg cgctctgggt cgcctggctg 840 tggaatgacc ctgttctcag gaagaagtac ccgggtgtca tctacgtccc tgagccctgg 900 gctttctaca cccttcacgt cagcagtcgg cactga 936 CLN6 amino acid sequence : (SEP ID NO: 4)
Met Glu Ala Thr Arg Arg Arg Gin His Leu Gly Ala Thr Gly Gly Pro 1 5 10 15
Gly Ala Gin Leu Gly Ala Ser Phe Leu Gin Ala Arg His Gly Ser Val
20 25 30
Ser Ala Asp Glu Ala Ala Arg Thr Ala Pro Phe His Leu Asp Leu Trp
35 40 45
Phe Tyr Phe Thr Leu Gin Asn Trp Val Leu Asp Phe Gly Arg Pro lie
50 55 60
Ala Met Leu Val Phe Pro Leu Glu Trp Phe Pro Leu Asn Lys Pro Ser 65 70 75 80
Val Gly Asp Tyr Phe His Met Ala Tyr Asn Val lie Thr Pro Phe Leu
85 90 95
Leu Leu Lys Leu He Glu Arg Ser Pro Arg Thr Leu Pro Arg Ser He
100 105 110
Thr Tyr Val Ser He He He Phe He Met Gly Ala Ser He His Leu
115 120 125
Val Gly Asp Ser Val Asn His Arg Leu Leu Phe Ser Gly Tyr Gin His
130 135 140
His Leu Ser Val Arg Glu Asn Pro He He Lys Asn Leu Lys Pro Glu
145 150 155 160
Thr Leu He Asp Ser Phe Glu Leu Leu Tyr Tyr Tyr Asp Glu Tyr Leu
165 170 175
Gly His Cys Met Trp Tyr He Pro Phe Phe Leu He Leu Phe Met Tyr
180 185 190
Phe Ser Gly Cys Phe Thr Ala Ser Lys Ala Glu Ser Leu He Pro Gly
195 200 205
Pro Ala Leu Leu Leu Val Ala Pro Ser Gly Leu Tyr Tyr Trp Tyr Leu
210 215 220
Val Thr Glu Gly Gin He Phe He Leu Phe He Phe Thr Phe Phe Ala
225 230 235 240
Met Leu Ala Leu Val Leu His Gin Lys Arg Lys Arg Leu Phe Leu Asp
245 250 255
Ser Asn Gly Leu Phe Leu Phe Ser Ser Phe Ala Leu Thr Leu Leu Leu
260 265 270
Val Ala Leu Trp Val Ala Trp Leu Trp Asn Asp Pro Val Leu Arg Lys
275 280 285
Lys Tyr Pro Gly Val He Tyr Val Pro Glu Pro Trp Ala Phe Tyr Thr
290 295 300
Leu His Val Ser Ser Arg His
305 310
CLN8 nucleotide sequence (SEQ ID NO: 5)
atgaatcctg cgagcgatgg gggcacatca gagagcattt ttgacctgga ctatgcatcc 60 tgggggatcc gctccacgct gatggtcgct ggctttgtct tctacttggg cgtctttgtg 120 gtctgccacc agctgtcctc ttccctgaat gccacttacc gttctttggt ggccagagag 180 aaggtcttct gggacctggc ggccacgcgt gcagtctttg gtgttcagag cacagccgca 240 ggcctgtggg ctctgctggg ggaccctgtg ctgcatgccg acaaggcgcg tggccagcag 300 aactggtgct ggtttcacat cacgacagca acgggattct tttgctttga aaatgttgca 360 gtccacctgt ccaacttgat cttccggaca tttgacttgt ttctggttat ccaccatctc 420 tttgcctttc ttgggtttct tggctgcttg gtcaatctcc aagctggcca ctatctagct 480 atgaccacgt tgctcctgga gatgagcacg ccctttacct gcgtttcctg gatgctctta 540 aaggcgggct ggtccgagtc tctgttttgg aagctcaacc agtggctgat gattcacatg 600 tttcactgcc gcatggttct aacctaccac atgtggtggg tgtgtttctg gcactgggac 660 ggcctggtca gcagcctgta tctgcctcat ttgacactgt tccttgtcgg actggctctg 720 cttacgctaa tcattaatcc atattggacc cataagaaga ctcagcagct tctcaatccg 780 gtggactgga acttcgcaca gccagaagcc aagagcaggc cagaaggcaa cgggcagctg 840 ctgcggaaga agaggccata g 861
CLN8 amino acid sequence (SEQ ID NO: 6)
Met Asn Pro Ala Ser Asp Gly Gly Thr Ser Glu Ser lie Phe Asp Leu
1 5 10 15
Asp Tyr Ala Ser Trp Gly lie Arg Ser Thr Leu Met Val Ala Gly Phe
20 25 30
Val Phe Tyr Leu Gly Val Phe Val Val Cys His Gin Leu Ser Ser Ser
35 40 45
Leu Asn Ala Thr Tyr Arg Ser Leu Val Ala Arg Glu Lys Val Phe Trp
50 55 60
Asp Leu Ala Ala Thr Arg Ala Val Phe Gly Val Gin Ser Thr Ala Ala
65 70 75 80
Gly Leu Trp Ala Leu Leu Gly Asp Pro Val Leu His Ala Asp Lys Ala
85 90 95
Arg Gly Gin Gin Asn Trp Cys Trp Phe His lie Thr Thr Ala Thr Gly
100 105 110
Phe Phe Cys Phe Glu Asn Val Ala Val His Leu Ser Asn Leu He Phe
115 120 125
Arg Thr Phe Asp Leu Phe Leu Val He His His Leu Phe Ala Phe Leu
130 135 140
Gly Phe Leu Gly Cys Leu Val Asn Leu Gin Ala Gly His Tyr Leu Ala 145 150 155 160
Met Thr Thr Leu Leu Leu Glu Met Ser Thr Pro Phe Thr Cys Val Ser
165 170 175
Trp Met Leu Leu Lys Ala Gly Trp Ser Glu Ser Leu Phe Trp Lys Leu
180 185 190
Asn Gin Trp Leu Met He His Met Phe His Cys Arg Met Val Leu Thr
195 200 205
Tyr His Met Trp Trp Val Cys Phe Trp His Trp Asp Gly Leu Val Ser
210 215 220
Ser Leu Tyr Leu Pro His Leu Thr Leu Phe Leu Val Gly Leu Ala Leu 225 230 235 240
Leu Thr Leu He He Asn Pro Tyr Trp Thr His Lys Lys Thr Gin Gin
245 250 255
Leu Leu Asn Pro Val Asp Trp Asn Phe Ala Gin Pro Glu Ala Lys Ser
260 265 270
Arg Pro Glu Gly Asn Gly Gin Leu Leu Arg Lys Lys Arg Pro
275 280 285
CB promoter (SEP ID NO: 71
ccacgttctg cttcactctc cccatctccc ccccctcccc acccccaatt ttgtatttat 60 ttatttttta attattttgt gcagcgatgg gggcgggggg gggggggggg cgcgcgccag 120 gcggggcggg gcggggcgag gggcggggcg gggcgaggcg gagaggtgcg gcggcagcca 180 atcagagcgg cgcgctccga aagtttcctt ttatggcgag gcggcggcgg cggcggccct 240 ataaaaagcg aagcgcgcgg cgggcgggag 270
CMV promoter (SEP ID NO: 81
cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc cccgcccatt 60 gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc attgacgtca 120 atgggtggag tatttacggt aaactgccca cttggcagta catcaagtgt atcatatgcc 180 aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt atgcccagta 240 catgacctta tgggactttc ctacttggca gtacatctac 280
P546 Promoter (SEP ID NO: 91
gaacaacgccaggctcctcaacaggcaactttgctacttctacagaaaatgataataaag
aaatgctggtgaagtcaaatgcttatcacaatggtgaactactcagcagggaggctctaa
taggcgccaagagcctagacttccttaagcgccagagtccacaagggcccagttaatcct
caacattcaaatgctgcccacaaaaccagcccctctgtgccctagccgcctcttttttcc
aagtgacagtagaactccaccaatccgcagctgaatggggtccgcctcttttccctgcct
aaacagacaggaactcctgccaattgagggcgtcaccgctaaggctccgccccagcctgg
gctccacaaccaatgaagggtaatctcgacaaagagcaaggggtggggcgcgggcgcgca
ggtgcagcagcacacaggctggtcgggagggcggggcgcgacgtctgccgtgcggggtcc
cggcatcggttgcgcgcgcgctccctcctctcggagagagggctgtggtaaaacccgtcc
ggaaaa

Claims

Claims What is claimed:
1. A composition comprising a gene therapy vector and a glycoside hydrolase enzyme.
2. The composition of claim 1 , wherein the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
3. The composition of claim 1 or 2, wherein the gene therapy vector is AAV8, AA9 or
Anc80.
4. The composition of any one of claims 1-3, wherein the composition is formulated for local intravenous delivery, sub-retinal delivery, intravitreal delivery or intrathecal delivery.
5. The composition of any one of claims 1-4, wherein the gene therapy vector and the glycoside hydrolase enzyme are admixed for administration simultaneously.
6. A kit comprising for delivering a transgene to a retinal cell of a subject comprising a a gene therapy vector and a glycoside hydrolase enzyme.
7. The kit of claim 6, wherein the the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
8. The kit of claim 6 or 7, wherein the gene therapy vector is AAV8, AA9 or Anc80.
9. A method of delivering a transgene to a retinal cell in a subject comprising administering i) a gene therapy vector encoding the transgene, and ii) a glycoside hydrolase enzyme to the subject.
10. A method of treating visual impairment or a vision-related disorder in a subject comprising administering i) a gene therapy vector encoding the transgene, and ii) a glycoside hydrolase enzyme to the subject, wherein delivery of the transgene effectively treats the visual impairment or the vision-related disorder.
11. The method of claim 9 or 10, wherein the glycoside hydrolase enzyme is
neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
12. The method of any one of claims 9-11, wherein the retinal cell is a bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell or amacrine cell.
13. The method of any one of claims 9-12, wherein the gene therapy vector is administered to the subject using local intravenous delivery, sub-retinal delivery, intravitreous delivery or intrathecal delivery.
14. The method of any one of claims 9-13, wherein the gene therapy vector encoding the transgene and the glycoside hydrolase enzyme are administered simultaneously to the subject.
15. The method of claim 14, wherein the gene therapy vector and the glycoside hydrolase enzyme are admixed.
16. The method of any one of claims 9-14, wherein the gene therapy vector and the glycoside hydrolase enzyme are administered separately.
17. The method of any one of claims 10-16 wherein the vision-related disorder is Batten disease, congenital cataracts, congenital glaucoma, retinal degeneration, optic atrophy, eye malformations. Strabismus, ocular misalignment, glaucoma, wet age-related macular degeneration, dry age-related macular degeneration, retinitis pigmentosa, choroideremia, Leber congenital amaurosis, Leber’s hereditary optic neuropathy, early onset retinal dystrophy, achromatopsia, x- linked retinoschisis, Usher Syndrome IB, neovascular age-related macular degeneration, Stargardt’s macular degeneration, diabetic macular degeneration, or diabetic macular edema.
18. A composition for delivering a transgene to a retinal cell in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene, and ii) a glycoside hydrolase enzyme.
19. A composition for treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene, and ii) a glycoside hydrolase enzyme.
20. A composition for delivering a transgene to a retinal cell in a subject, wherein the composition comprises a gene therapy vector encoding the transgene, wherein the composition is administered with a second composition comprising a glycoside hydrolase enzyme.
21. A composition for treating a treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises a gene therapy vector encoding the transgene, wherein the composition is administered with a second composition comprising a glycoside hydrolase enzyme.
22. A composition for delivering a transgene to a retinal cell in a subject, wherein the composition comprises a glycoside hydrolase enzyme, wherein the composition is administered with a second composition comprising a gene therapy vector encoding the transgene.
23. A composition for treating a treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises a glycoside hydrolase enzyme, wherein the composition is administered with a second composition comprising a gene therapy vector encoding the transgene.
24. The composition of any one of claims 19-23, wherein the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
25. The composition of any one of claims 19-24, wherein the retinal cell is a bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell or amacrine cell.
26. The composition of any one of claims 19-25, wherein the composition is formulated for local intravenous delivery, sub-retinal delivery, intravitreal delivery or intrathecal delivery.
27. The composition of any one of claims 20-26, wherein the composition and the second composition are administered simultaneously to the subject.
28. The composition of any one of claims 20-27, wherein the composition and the second composition are admixed.
29. The composition of any one of claims 20-27, wherein the composition and the second composition are administered separately.
30. The composition of any one of claims 19, 21 or 23-29, wherein the vision-related disorder is Batten disease, congenital cataracts, congenital glaucoma, retinal degeneration, optic atrophy, eye malformations. Strabismus, ocular misalignment, glaucoma, wet age-related macular degeneration, dry age-related macular degeneration, retinitis pigmentosa, choroideremia, Leber congenital amaurosis, Leber’s hereditary optic neuropathy, early onset retinal dystrophy, achromatopsia, x-linked retinoschisis, Usher Syndrome IB, neovascular age-related macular degeneration, Stargardt’s macular degeneration, diabetic macular degeneration, or diabetic macular edema.
31. Use of composition for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene, and ii) a glycoside hydrolase enzyme.
32. Use of a composition for the preparation of a medicament for treating visual impairment or a vision-related disorder in a subject, wherein the composition comprises i) a gene therapy vector encoding the transgene, and ii) a glycoside hydrolase enzyme.
33. Use of a gene therapy vector encoding a transgene for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the medicament is administered with a composition comprising a glycoside hydrolase enzyme.
34. Use of a gene therapy vector encoding a transgene for the preparation of a medicament for treating visual impairment or a vision-related disorder in a subject, wherein the medicament is administered with a composition comprising a glycoside hydrolase enzyme.
35. Use of glycoside hydrolase enzyme for the preparation of a medicament for delivering a transgene to a retinal cell in a subject, wherein the medicament is administered with a composition comprising a gene therapy vector encoding the transgene.
36. Use of glycoside hydrolase enzyme for the preparation of a medicament for treating a treating visual impairment or a vision-related disorder in a subject, wherein the medicament is administered with a composition comprising a gene therapy vector encoding the transgene.
37. The use of any one of claims 32-36, wherein the glycoside hydrolase enzyme is neuraminidase, lactase, amylase, chitinase, cellulase, sucrase, maltase, invertase, or lysozyme.
38. The use of any one of claims 32-37, wherein the retinal cell is a bipolar cell, rod photoreceptor cell, cone photoreceptor cell, ganglion cell, Mueller glia cell, microglia cell, horizontal cell or amacrine cell.
39. The use of any one of claims 32-38, wherein the medicament is formulated for local intravenous delivery, sub-retinal delivery, intravitreal delivery or intrathecal delivery.
40. The use of any one of claims 33-39, wherein the medicament and the composition are administered simultaneously to the subject.
41. The use of any one of claims 33-39, wherein the composition and the composition are admixed.
42. The use of any one of claims 33-39, wherein the medicament and the composition are administered separately.
43. The use of any one of claims 32, 34 or 36-42, wherein the vision-related disorder is Batten disease, congenital cataracts, congenital glaucoma, retinal degeneration, optic atrophy, eye malformations. Strabismus, ocular misalignment, glaucoma, wet age-related macular degeneration, dry age-related macular degeneration, retinitis pigmentosa, choroideremia, Leber congenital amaurosis, Leber’s hereditary optic neuropathy, early onset retinal dystrophy, achromatopsia, x- linked retinoschisis, Usher Syndrome IB, neovascular age-related macular degeneration, Stargardt’s macular degeneration, diabetic macular degeneration, or diabetic macular edema.
PCT/US2020/028207 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme WO2020236351A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2021568543A JP2022533645A (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using glycoside hydrolase enzymes
EP20723738.9A EP3969059A1 (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme
CN202080051735.0A CN114126667A (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using glycoside hydrolases
CA3141017A CA3141017A1 (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme
US17/611,972 US20220233655A1 (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme
KR1020217040926A KR20220009427A (en) 2019-05-17 2020-04-15 Improved Delivery of Gene Therapy Vectors to Retinal Cells Using Glycoside Hydrolases
AU2020278499A AU2020278499A1 (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962849794P 2019-05-17 2019-05-17
US62/849,794 2019-05-17

Publications (1)

Publication Number Publication Date
WO2020236351A1 true WO2020236351A1 (en) 2020-11-26

Family

ID=70482910

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2020/028207 WO2020236351A1 (en) 2019-05-17 2020-04-15 Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme
PCT/US2020/028352 WO2020236352A1 (en) 2019-05-17 2020-04-15 Optimized gene therapy targeting retinal cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2020/028352 WO2020236352A1 (en) 2019-05-17 2020-04-15 Optimized gene therapy targeting retinal cells

Country Status (10)

Country Link
US (2) US20220226507A1 (en)
EP (2) EP3969060A1 (en)
JP (2) JP2022533645A (en)
KR (1) KR20220009427A (en)
CN (1) CN114126667A (en)
AR (1) AR118696A1 (en)
AU (2) AU2020278499A1 (en)
CA (2) CA3141017A1 (en)
TW (1) TW202110486A (en)
WO (2) WO2020236351A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114516901B (en) * 2022-03-11 2023-01-17 上海勉亦生物科技有限公司 AAV vector with high affinity for nervous system and application thereof
CN115323001A (en) * 2022-07-28 2022-11-11 深圳先进技术研究院 Gene delivery system for targeting retina and application thereof

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1995013392A1 (en) 1993-11-09 1995-05-18 Medical College Of Ohio Stable cell lines capable of expressing the adeno-associated virus replication gene
WO1995013365A1 (en) 1993-11-09 1995-05-18 Targeted Genetics Corporation Generation of high titers of recombinant aav vectors
WO1996017947A1 (en) 1994-12-06 1996-06-13 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant aav vectors
WO1997006243A1 (en) 1995-08-10 1997-02-20 Pasteur Merieux Serums Et Vaccins Method for purifying viruses by chromatography
WO1997008298A1 (en) 1995-08-30 1997-03-06 Genzyme Corporation Chromatographic purification of adenovirus and aav
WO1997009441A2 (en) 1995-09-08 1997-03-13 Genzyme Corporation Improved aav vectors for gene therapy
WO1997021825A1 (en) 1995-12-15 1997-06-19 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
WO1998009657A2 (en) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Method for recombinant adeno-associated virus-directed gene therapy
US5786211A (en) 1994-06-06 1998-07-28 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5871982A (en) 1994-10-28 1999-02-16 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV virus and methods of use thereof
WO1999011764A2 (en) 1997-09-05 1999-03-11 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of recombinant aav vectors
US6258595B1 (en) 1999-03-18 2001-07-10 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
WO2001083692A2 (en) 2000-04-28 2001-11-08 The Trustees Of The University Of Pennsylvania Recombinant aav vectors with aav5 capsids and aav5 vectors pseudotyped in heterologous capsids
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US20050053922A1 (en) 2003-06-30 2005-03-10 Schaffer David V. Mutant adeno-associated virus virions and methods of use thereof
US20090202490A1 (en) 2003-06-30 2009-08-13 Schaffer David V Mutant adeno-associated virus virions and methods of use thereof
WO2015128624A1 (en) * 2014-02-25 2015-09-03 The University Of Manchester Treatment of retinal degeneration using gene therapy
US9434928B2 (en) 2011-11-23 2016-09-06 Nationwide Children's Hospital, Inc. Recombinant adeno-associated virus delivery of alpha-sarcoglycan polynucleotides
US9613872B2 (en) 2014-09-29 2017-04-04 Kabushiki Kaisha Toshiba Method of manufacturing semiconductor device
US9614423B2 (en) 2012-04-07 2017-04-04 Traugott Weller Method for producing rotating electrical machines
US9620777B2 (en) 2013-09-30 2017-04-11 Tdk Corporation Positive electrode and lithium ion secondary battery using thereof
US9818600B2 (en) 2014-03-21 2017-11-14 Hitachi Kokusai Electric, Inc. Substrate processing apparatus and method of manufacturing semiconductor device

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20170260A1 (en) * 2014-05-02 2017-04-12 Genzyme Corp VECTORS OF AAV FOR GENE THERAPY OF THE RETINA AND CNS

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1995013392A1 (en) 1993-11-09 1995-05-18 Medical College Of Ohio Stable cell lines capable of expressing the adeno-associated virus replication gene
WO1995013365A1 (en) 1993-11-09 1995-05-18 Targeted Genetics Corporation Generation of high titers of recombinant aav vectors
US5658776A (en) 1993-11-09 1997-08-19 Targeted Genetics Corporation Generation of high titers of recombinant AAV vectors
US5786211A (en) 1994-06-06 1998-07-28 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5871982A (en) 1994-10-28 1999-02-16 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV virus and methods of use thereof
WO1996017947A1 (en) 1994-12-06 1996-06-13 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant aav vectors
WO1997006243A1 (en) 1995-08-10 1997-02-20 Pasteur Merieux Serums Et Vaccins Method for purifying viruses by chromatography
WO1997008298A1 (en) 1995-08-30 1997-03-06 Genzyme Corporation Chromatographic purification of adenovirus and aav
WO1997009441A2 (en) 1995-09-08 1997-03-13 Genzyme Corporation Improved aav vectors for gene therapy
WO1997021825A1 (en) 1995-12-15 1997-06-19 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
WO1998009657A2 (en) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Method for recombinant adeno-associated virus-directed gene therapy
WO1999011764A2 (en) 1997-09-05 1999-03-11 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of recombinant aav vectors
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6258595B1 (en) 1999-03-18 2001-07-10 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
WO2001083692A2 (en) 2000-04-28 2001-11-08 The Trustees Of The University Of Pennsylvania Recombinant aav vectors with aav5 capsids and aav5 vectors pseudotyped in heterologous capsids
US20050053922A1 (en) 2003-06-30 2005-03-10 Schaffer David V. Mutant adeno-associated virus virions and methods of use thereof
US20090202490A1 (en) 2003-06-30 2009-08-13 Schaffer David V Mutant adeno-associated virus virions and methods of use thereof
US9434928B2 (en) 2011-11-23 2016-09-06 Nationwide Children's Hospital, Inc. Recombinant adeno-associated virus delivery of alpha-sarcoglycan polynucleotides
US9614423B2 (en) 2012-04-07 2017-04-04 Traugott Weller Method for producing rotating electrical machines
US9620777B2 (en) 2013-09-30 2017-04-11 Tdk Corporation Positive electrode and lithium ion secondary battery using thereof
WO2015128624A1 (en) * 2014-02-25 2015-09-03 The University Of Manchester Treatment of retinal degeneration using gene therapy
US9818600B2 (en) 2014-03-21 2017-11-14 Hitachi Kokusai Electric, Inc. Substrate processing apparatus and method of manufacturing semiconductor device
US9613872B2 (en) 2014-09-29 2017-04-04 Kabushiki Kaisha Toshiba Method of manufacturing semiconductor device

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
ARSOV, T.SMITH, K. R.DAMIANO, J.FRANCESCHETTI, S.CANAFOGLIA, L.BROMHEAD, C. J.BERKOVIC, S. F.: "Kufs disease, the major adult form of neuronal ceroid lipofuscinosis, caused by mutations in CLN6", AMERICAN JOURNAL OF HUMAN GENETICS, vol. 88, no. 5, 2011, pages 566 - 573, XP028206956, Retrieved from the Internet <URL:https://doi.org/10.1016/j.ajhg.2011.04.004> DOI: 10.1016/j.ajhg.2011.04.004
BRAS, J.VERLOES, A.SCHNEIDER, S. A.MOLE, S. E.GUERREIRO, R. J.: "Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis", HUMAN MOLECULAR GENETICS, vol. 27, no. 12, 2012, pages 2646 - 2650, Retrieved from the Internet <URL:https://doi.org/10.1093/hmg/dds089>
CANAFOGLIA, L.MORBIN, M.SCAIOLI, V.PAREYSON, D.D'INCERTI, L.FUGNANESI, VFRANCESCHETTI, S.: "Recurrent generalized seizures, visual loss, and palinopsia as phenotypic features of neuronal ceroid lipofuscinosis due to progranulin gene mutation", EPILEPSIA, vol. 55, no. 6, 2014, pages e56 - e59, XP055362995, Retrieved from the Internet <URL:https://doi.org/10.1111/epi.12632> DOI: 10.1111/epi.12632
CANNELLI, N.CASSANDRINI, D.BERTINI, E.STRIANO, P.FUSCO, L.GAGGERO, R.SANTORELLI, F. M.: "Novel mutations in CLN8 in Italian variant late infantile neuronal ceroid lipofuscinosis: another genetic hit in the Mediterranean", NEUROGENETICS, vol. 7, no. 2, 2006, pages 111 - 117, XP019385842, Retrieved from the Internet <URL:https://doi.org/10.1007/sl0048-005-0024-y> DOI: 10.1007/s10048-005-0024-y
CARTER, CURRENT OPINIONS IN BIOTECHNOLOGY, 1992, pages 1533 - 539
CHOUDHURY ET AL., MOL. THER., vol. 24, no. 7, 2016, pages 1247 - 1257
CLARK ET AL., GENE THERAPY, vol. 3, 1996, pages 1124 - 1132
CLARK ET AL., HUM. GENE THER., vol. 10, no. 6, 1999, pages 1031 - 1039
ESTRADA-CUZCANO, A.MARTIN, S.CHAMOVA, T.SYNOFZIK, M.TIMMANN, D.HOLEMANS, T.SCHULE, R.: "Loss-of-function mutations in the ATP13A2/PARK9 gene cause complicated hereditary spastic paraplegia (SPG78", BRAIN : A JOURNAL OF NEUROLOGY, vol. 140, no. 2, 2017, pages 287 - 305, Retrieved from the Internet <URL:https://doi.org/10.1093/brain/aww307>
GAO ET AL., J. VIROL., vol. 78, 2004, pages 6381 - 6388
GILES ET AL., MOLECULAR THERAPY, vol. 26, no. 12, 2018, pages 2848 - 2862
HERMONAT ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6466
J. CEHAJIC-KAPETANOVIC ET AL: "Glycosidic enzymes enhance retinal transduction following intravitreal delivery of AAV2", MOLECULAR VISION, 17, 30 June 2011 (2011-06-30), pages 1771 - 1783, XP055183839 *
KARALI ET AL., NUCLEIC ACIDS RES., vol. 44, no. 4, 29 February 2016 (2016-02-29), pages 1525 - 1540
KNIFFIN, C.: "OMIM Clinical Synopsis - %609055 - CEROID LIPOFUSCINOSIS", NEURONAL, vol. 9, 19 March 2019 (2019-03-19), Retrieved from the Internet <URL:http://omim.org/clinicalSynopsis/609055>
LAUGHLIN ET AL., GENE, vol. 23, 1983, pages 65 - 73
LEBKOWSKI ET AL., 1988 MOL. CELL. BIOL., vol. 7, 1988, pages 349
LEE ET AL., PROGRESS IN RETINAL AND EYE RESEARCH, vol. 68, 2019, pages 31 - 53
MARSHALL ET AL., NEUROLOGY, vol. 65, no. 2, 2005, pages 275 - 279
MARSIC ET AL., MOLECULAR THERAPY, vol. 22, no. 11, 2014, pages 1900 - 1909
MCLAUGHLIN ET AL., J. VIROL., vol. 62, 1988, pages 1963
MOEN, M. N.FJAER, R.HAMDANI, E. H.LAERDAHL, J. K.MENCHINI, R. J.VIGELAND, M. D.CHAUDHRY, F. A.: "Pathogenic variants in KCTD7 perturb neuronal K fluxes and glutamine transport", BRAIN, vol. 139, no. 12, 2016, pages 3109 - 3120, Retrieved from the Internet <URL:https://doi.org/10.1093/brain/aww244>
MOL. THER., vol. 13, no. 1, 2006, pages 67 - 76
MOLE, S. E.WILLIAMS, R. E.: "Neuronal Ceroid-Lipofuscinoses. GeneReviews®", 1993, UNIVERSITY OF WASHINGTON
MUZYCZKA, CURR. TOPICS IN MICROBIAL. AND IMMUNOL., vol. 158, 1992, pages 97 - 129
MUZYCZKA, CURRENT TOPICS IN MICROBIOLOGY AND IMMUNOLOGY, vol. 158, 1992, pages 97 - 129
NOSKOVA, L.STRANECKY, V.HARTMANNOVA, H.PRISTOUPILOVA, A.BARESOVA, V.IVANEK, R.KMOCH, S.: "Mutations in DNAJC5, encoding cysteine-string protein alpha, cause autosomal-dominant adult-onset neuronal ceroid lipofuscinosis", AMERICAN JOURNAL OF HUMAN GENETICS, vol. 89, no. 2, 2011, pages 241 - 252, Retrieved from the Internet <URL:https://doi.org/10.1016/j.ajhg.2011.07.003>
PAUL ET AL., HUMAN GENE THERAPY, vol. 4, 1993, pages 609 - 615
PERRIN ET AL., VACCINE, vol. 13, 1995, pages 1244 - 1250
RATSCHIN ET AL., MOL. CELL. BIOL., vol. 4, 1984, pages 2072
ROOSING, S.VAN DEN BORN, L. I.SANGERMANO, R.BANFI, S.KOENEKOOP, R. K.ZONNEVELD-VRIELING, M. N.HOYNG, C. B.: "Mutations in MFSD8, Encoding a Lysosomal Membrane Protein, Are Associated with Nonsyndromic Autosomal Recessive Macular Dystrophy", OPHTHALMOLOGY, vol. 122, no. 1, 2015, pages 170 - 179, Retrieved from the Internet <URL:https://doi.org/10.1016/j.ophtha.2014.07.040>
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
SAMULSKI ET AL., J. VIROL., vol. 63, 1989, pages 3822 - 3828
SAMULSKI ET AL., PROC. NATL. ACAD. S6. USA, vol. 79, 1982, pages 2077 - 2081
SCHENPPCLARK, METHODS MOL. MED., vol. 69, 2002, pages 427 - 443
SENAPATHYCARTER, J. BIOL. CHEM., vol. 259, 1984, pages 4661 - 4666
SHEN ET AL., J. BIOL. CHEM., vol. 286, 2011, pages 13532 - 13540
SRIVASTAVA ET AL., J. VIROL., vol. 45, 1983, pages 555 - 564
STAROPOLI, J. F.KARAA, A.LIM, E. T.KIRBY, A.ELBALALESY, N.ROMANSKY, S. G.COTMAN, S. L.: "A homozygous mutation in KCTD7 links neuronal ceroid lipofuscinosis to the ubiquitin-proteasome system", AMERICAN JOURNAL OF HUMAN GENETICS, vol. 97, no. 1, 2012, pages 202 - 208, XP028425647, Retrieved from the Internet <URL:https://doi.org/10.1016/j.ajhg.2012.05.023> DOI: 10.1016/j.ajhg.2012.05.023
TRAPANI IVANA ET AL: "Vector platforms for gene therapy of inherited retinopathies", PROGRESS IN RETINAL AND EYE RESEARCH, OXFORD, GB, vol. 43, 12 August 2014 (2014-08-12), pages 108 - 128, XP029075135, ISSN: 1350-9462, DOI: 10.1016/J.PRETEYERES.2014.08.001 *
TRATSCHIN ET AL., MOL. CELL. BIOL., vol. 5, 1985, pages 3251
VIROLOGY, vol. 330, no. 2, 2004, pages 375 - 383
WEIZMANN: "Ceroid Lipofuscinosis, Neuronal, 10 disease: Malacards - Research Articles, Drugs, Genes, Clinical Trials", 19 March 2019, INSTITUTE OF SCIENCE
XIN, W.MULLEN, T. E.KIELY, R.MIN, J.FENG, X.CAO, Y.SIMS, K.: "CLN5 mutations are frequent in juvenile and late-onset non-Finnish patients with NCL", NEUROLOGY, vol. 74, no. 7, 2010, pages 565 - 571
ZINN ET AL., CELL REPORTS, vol. 12, 2015, pages 1056 - 1068

Also Published As

Publication number Publication date
US20220226507A1 (en) 2022-07-21
EP3969059A1 (en) 2022-03-23
AU2020278960A1 (en) 2021-12-23
CA3141020A1 (en) 2020-11-26
CA3141017A1 (en) 2020-11-26
TW202110486A (en) 2021-03-16
AR118696A1 (en) 2021-10-27
US20220233655A1 (en) 2022-07-28
CN114126667A (en) 2022-03-01
AU2020278499A1 (en) 2022-01-06
WO2020236352A1 (en) 2020-11-26
JP2022533983A (en) 2022-07-27
EP3969060A1 (en) 2022-03-23
JP2022533645A (en) 2022-07-25
KR20220009427A (en) 2022-01-24

Similar Documents

Publication Publication Date Title
EP2699270B1 (en) Adeno-associated virus virions with variant capsid and methods of use thereof
US10849991B2 (en) Gene therapy for the treatment of a disease of retinal cone cells
US20220233655A1 (en) Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme
CN114144518A (en) Dual leucine zipper kinase inhibitors for gene therapy
AU2020366242A1 (en) Materials and methods for the treatment of disorders associated mutations in the IRF2BPL gene
WO2021102435A1 (en) Materials and methods for treatment of disorders associated with the ighmbp2 gene
AU2022256458A1 (en) Recombinant adeno-associated virus encoding methyl-cpg binding protein 2 for treating pitt hopkins syndrome via intrathecal delivery
WO2024011115A1 (en) Adeno-associated virus delivery of cln1 polynucleotide
EP3807309A1 (en) Recombinant adeno-associated virus products and methods for treating dystroglycanopathies and laminin-deficient muscular dystrophies
CA3238030A1 (en) Materials and methods for slc6a1 gene therapy
JP2022523050A (en) Highly efficient transduction and lateral spread in the retina by a novel AAV virus enhanced by rational design

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20723738

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021568543

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3141017

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20217040926

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020278499

Country of ref document: AU

Date of ref document: 20200415

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020723738

Country of ref document: EP

Effective date: 20211217